在异种移植中TF对IBMIR的启动作用及人Treg对移植物保护作用的研究
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:临床上胰岛移植的应用给I型糖尿病患者带来了希望,同时猪的器官在器官移植中的应用解决了捐赠器官严重短缺的问题。但是,无论是同种还是异种胰岛经门静脉移植,都面临着移植后早期快速的大量胰岛丢失的问题以及受体体内产生的免疫排斥反应最终导致移植物排斥。因此本研究:①利用siRNA对新生猪胰岛细胞团的TF目标基因进行特异性地表达沉默,分析TF对NICC与血液接触后发生的IBMIR的始动作用,探寻一种更为安全有效的措施来抑制IBMIR,减少胰岛的丢失提高胰岛的植入率;②应用NOD-SCIDIL2rγ-/-小鼠作为猪胰岛移植受体,研究注射人外周血单个细胞和体外扩增的nTregs后是否可以诱导以及如何诱导对胰岛移植物的免疫耐受,建立一种以经体外扩增培养的Tregs为基础的治疗方法用以抑制T细胞介导的异种移植排斥反应,从而减轻或消除免疫抑制治疗的需求,为人nTregs在临床移植治疗的实际应用提供了参考。
     方法和结果:
     1.新生猪胰岛细胞组织因子基因沉默
     本研究通过在新生猪胰腺原位及分离培养后的NICC免疫组织化学检测均可见TF呈阳性染色,从而证实猪的胰岛细胞自身就有TF的表达而不是因为细胞分离过程所导致的TF表达。本研究运用siRNA在基因水平上阻断猪胰岛细胞TF的表达,以抑制细胞与血液接触后触发的IBMIR,从而减少猪胰岛的丢失。实验设计了5条siRNA,单独或两两组合或三三组合按最佳转染条件分别转染NICC,结果发现其中三条siRNA组合可以使NICC的TF基因沉默效率达53%-70%,平均60%,p<0.01,且TF蛋白表达水平下降近50%。同时流式细胞仪检测结果显示,siRNA转染后,NICC的活性未受明显的影响,排除了因细胞死亡所导致NICC蛋白表达水平的降低。上述结果提示,设计的5条siRNA中有3条siRNA组合可显著降低NICC在基因及蛋白水平TF的表达,以便于进一步研究TF在IBMIR发生中的作用。
     2.新生猪胰岛细胞组织因子基因沉默体外抑制IBMIR
     本研究将未转染siRNA的NICC,转染对照siRNA的NICC和转染TF siRNA的NICC与人外周血在体外tubing loops模型内共孵育60分钟后,观察血凝块的大小及重量,血细胞计数检测血细胞的消耗,ELISA检测血浆中TAT及C3a的浓度,免疫组织化学检测中性粒细胞浸润及IgG沉积情况,Real-time PCR检测各组NICC的α-Gal基因表达。结果显示,转染TFsiRNA的NICC与人外周血共孵育后,不仅其血凝块的形成较其他两组受到明显的抑制,血凝块的大小和体积均有明显减小,而且血细胞计数显示血液中血小板、白细胞和中性粒细胞的消耗显著减少,补体激活受到明显的抑制。结果还显示,TF在NICC上的表达受到抑制后,血浆中TAT的浓度显著降低,中性粒细胞的浸润明显减少。上述结果提示阻断TF在胰岛上的表达可以抑制凝血、补体激活和前炎症介质的产生,也就抑制了IBMIR的发生。同时免疫荧光染色结果显示,NICC的TF表达降低并不能明显减轻血凝块中IgG在NICC周围的沉积,Real-time PCR结果表明TF特异性的siRNA对NICC TF基因的沉默作用没有影响到细胞a-Gal基因的表达,提示抗体介导的凝血激活通路并不是体外Loops模型中IBMIR发生的主要启动因子。
     3.人源化小鼠模型研究人调节性T细胞介导的异种胰岛移植耐受
     本研究在体外用CD4+CD25+CD 127dim/- Treg分离试剂盒从健康自愿者外周血单个核细胞分离获得CD4+CD25+调节性T细胞,应用IL-2,抗CD3/CD28抗体磁珠和雷帕霉素体外扩增纯化CD4+CD25+ Tregs,扩增后的Treg经流式细胞仪检测具有典型Treg的表型,其中CD4+CD25+Foxp3+的细胞达到90%以上;同时在体外检测扩增后的Treg的抑制功能。混合淋巴细胞反应(mixed lymphocyte reaction,MLR)的结果显示,体外扩增的Tregs可以明显抑制针对猪异种抗原的效应性T细胞的增殖效应;收集MLR各孔细胞培养上清并检测各种细胞因子的浓度,结果发现体外扩增的Tregs可显著抑制效应性T细胞分泌效应性细胞因子如IL-2和[FN-γ,而明显促进抑制性细胞因子IL-10的分泌,但细胞培养上清中TGF-β的浓度没有明显的变化。上述结果提示,经体外扩增体系扩增的Tregs具备调节性T细胞的表型和抑制功能。本研究体内实验是将分离培养的NICC移植至NOD-SCIDIL2rγ-/-受体小鼠的肾被膜下,于移植后的7天内,将1×107去除CD25+细胞的人PBMC或PBMC与2×106体外扩增的nTregs共同经尾静脉注射给接受NICC移植的NOD-SCID IL2rγ-/-小鼠体内。免疫组化用以检测移植物的存活情况,流式细胞仪检测人细胞植入情况,FACS和Real-time PCR分析移植耐受或排斥小鼠体内人nTregs的表型和基因表达。结果显示,仅接受NICC移植的NOD-SCID IL2rγ-/-小鼠,移植物可存活100天以上,免疫组化检测结果显示移植胰岛的胰岛素,胰高血糖素和生长抑素染色呈阳性,说明NOD-SCID IL2rγ-/-小鼠可以耐受猪胰岛移植物。NOD-SCID IL2rγ-/-受体小鼠静脉接受1×107去除CD25+细胞的人PBMC后,28天内免疫组化检测NICC移植物几乎被完全排斥,同时有较多的细胞浸润在移植部位,这些浸润的细胞经免疫组化进一步检测绝大部分为人CD45+细胞,其中包括CD4+及CD8+细胞,免疫荧光双染仅见CD4+细胞,未见CD4+Foxp3+细胞,流式细胞仪检测移植物浸润细胞表型的结果同样显示浸润的CD4+细胞中几乎没有CD25+Foxp3+双阳性细胞;而同时接受1×107去除CD25+细胞的人PBMC与2×106体外扩增的nTregs(?)植入的宿主小鼠NICC移植物却未被排斥,其存活时间不仅延长至100天以上,而且具有分泌胰岛素,胰高血糖素和生长抑素的功能,HE染色显示细胞植入28天可见有细胞围绕在胰岛的周围,但未浸润入胰岛中。这些围绕在胰岛周围的细胞经过免疫组化检测绝大部分亦为人CD45+细胞,其中包括CD4+及CD8+细胞,但免疫荧光双染结果显示在CD4+阳性细胞中,可见CD4+Foxp3+双阳性细胞。流式细胞仪检测移植物周围的细胞表型结果显示浸润的CD4+细胞中有高达18.6±2.3%的CD25+Foxp3+双阳性细胞。上述结果提示移植NICC的NOD-SCID IL2rγ-/-受体小鼠静脉接受1×107去除CD25+细胞的人PBMC后,人PBMC即可对猪异种抗原产生免疫应答而导致NICC移植物被排斥,参与免疫排斥的细胞主要包括CD4+和CD8+T细胞;如果同时植入体外扩增的自体2×106Tregs后,NICC移植物的排斥延迟,Treg抑制了自体PBMC对猪异种抗原产生的免疫应答而保护了NICC移植物。Real-time PCR检测NOD-SCID IL2rγ-/-受体小鼠移植部位浸润细胞基因表达水平结果同样显示Treg与PBMC共植入小鼠的Foxp3,CTLA-4及IL-10的基因表达水平明显高于仅接受PBMC植入小鼠的表达水平,p<0.01,但IFN-γ的表达水平却显著低于仅接受PBMC植入小鼠的表达水平,p<0.01;受体小鼠血清各种细胞因子浓度的检测结果显示,Treg与PBMC共植入小鼠血清中的IL-10的浓度明显高于仅PBMC植入小鼠血清IL-10的浓度,p<0.01,而IFN-γ的表达水平显著低于仅PBMC植入小鼠的表达水平,p<0.01。上述结果提示,Treg与PBMC共植入小鼠体内,Tregs可通过抑制效应性细胞因子IFN-y的表达,促进抑制性细胞因子IL-10的表达,从而抑制了人PBMC针对猪异种抗原产生的免疫排斥反应。
     结论:本研究应用TF siRNA特异性地抑制了NICC TF的表达,NICC TF的基因沉默能在体外抑制IBMIR的发生;人Tregs可抑制人PBMCs(去除CD25+细胞)介导的异种免疫排斥反应而保护NOD-SCID IL2rg-/-受体小鼠NICC移植物,且Tregs在体内的抑制功能与IL-10参与有关。拟人化NOD-SCID IL2rg-/-小鼠在胰岛异种移植中人Tregs介导的免疫耐受及其机制的研究中是一种非常合适的模型。
Clinical islet transplantation is a promising treatment for typeⅠdiabetes and using pig organs could solve the significant increasing shortage of donor organs for allotransplantation. However early innate immune destruction of islets and rejection are major problems preventing clinical trials. Therefore in this study,①we used small interfering RNAs (siRNAs)-mediated gene knockdown to specifically silence TF gene expression in neonatal porcine islet cell clusters (NICC). This gene targeting approach allowed us to not only analyze directly the importance of TF in the initiation of IBMIR mediated by NICC, but also to explore the potential targets for developing strategies that can enhance islet engraftment and survival;②Using NOD-SCID IL2rγ-/- mice as recipients for pig islet transplantation, we studied whether and how immune tolerance of islet grafts to T-cell mediated cellular rejection is induced by reconstitution of these mice with human PBMCs and nTregs and produced a cell-based therapy that can be developed ex vivo that will suppress the T cell mediated response to the xeno-graft and thereby reduce or eliminate the requirement for immunosuppression.
     1. TF knockdown in NICC
     TF is expressed on newly isolated NICC and can be detected on neonatal pig pancreas tissue prior to NICC production confirming that it is not a by product of the isolation procedure. In order to test the hypothesis that specific suppression of islet TF expression would lead to a significant inhibition of IBMIR, small interfering RNA (siRNA) was used to knockdown TF gene expression in NICC. 5 pairs of siRNA were designed and transfected into NICC respectively or with different combinations under the optimal conditions. A 60%(50%-70%) and a 50% reduction in TF gene and protein expression respectively was achieved with selected 3 pairs of siRNA. FACS results showed that siRNA transfection had no significant effect on the viability of NICC used in these experiments. These data suggested that siRNA transfection was sufficient to specifically knockdown TF gene and protein expression in NICC.
     2. Suppressed TF expression in NICC led to inhibition of IBMIR in vitro
     After transfection, non-transfected, control siRNA transfected and TF siRNA transfected NICC were cultured with ABO compatible human blood in loops tubing respectively for 60 mins. Clot size and weight and blood cell counts were determined. Plasma was analyzed to determine levels of thrombin-antithrombin complexes (TAT) and C3a using commercially available ELISA kits. Immunohistochemical staining for neutrophil elastase and human IgG were used to determine neutrophil infiltration and antibody deposit in clots.α-Gal gene expression as assessed by real-time PCR. The results showed that TF siRNA resulted in substantially reduced not only clot size and weight but also the consumption of platelets, white blood cells and neutrophils. Consistent with these findings TAT and C3a levels were significantly less in loops containing TF siRNA transfected NICC compared to those with non-transfected or control siRNA transfected NICC. When TF siRNA transfected NICC were included in the loops with human blood, the number of infiltrating neutrophils within the clot was decreased considerably. These data showed that TF expression was an important initiator of the NICC mediated inflammatory response and this response was inhibited by TF knockdown. Meantime immunofluorescence staining of NICC containing clots taken from the loops showed strong deposition of human IgG which was not diminished when TF siRNA transfected NICC were used and a-Gal gene expression as assessed by real-time PCR was similar in all three groups. These data suggested that antibody mediated activation of the alternative pathway of thrombosis was not the major initiating event in this study.
     3. Islet xenograft protection by ex vivo-expanded human regulatory T cells in humanized NOD-SCID IL2rγ-/- mice
     CD4+CD25+CD127 dim/- T cells were isolated from PBMC by using a CD4+CD25+CD127 dim/- regulatory T cell isolation kit. Fresh Treg cells were cultured with human IL-2, CD3/CD28 Dynabeads and rapamycin for expansion. The expanded cells had the typical phenotype of Tregs and the percentage of CD4+CD25+Foxp3+ triple positive cells was over 90%; Mixed lymphocyte reaction (MLR) was used to investigate the suppression function of expanded Tregs. The results showed that expanded human Treg cells were able to suppress antiporcine proliferative responses in vitro. The concentration of IL-2 and IFN-γwere reduced and the IL-10 level was upregulated in the co-cultured cells supernatant, but the concentration of TGF-βhad no change. These data demonstrated that expanded Treg cells retained the phenotype and suppressive activity. NICC were transplanted to NOD-SCID IL2rγ-/1 mice under renal capsule.1×10'CD25+cell-depleted human PBMCs were co-transferred with or without 2×106 expanded human nTregs into the NICC recipient NOD-SCID IL2rγ-/- mice within 7 days after transplantation. Graft survival was monitored by histology. Human cell engraftment was determined by FACS. Phenotype of CD25-human cells and expanded human Tregs and their gene expression were analysed by FACS and real-time PCR. The results showed that NICC xenograft was intact with insulin, glucogan and somatostatin positive staining in NOD-SCID IL2rγ-/- recipient mice without human cell transfer for at least 100 days after transplantation. While NICC grafts were rejected in NOD-SCID IL2rγ-/- recipient mice 28 days after transfer with 1x107 human CD25+ cell-depleted PBMCs in the absence of autologous Tregs. There were lots of cells infiltrated in the graft which were CD45+ cells including CD4+ and CD8+ cells detected by immunohistology and only CD4 positive staining cells were detected by immunofluorescence double staining. NICC graft infiltrating human CD4+ cells isolated from rejecting NOD-SCID IL2rg-/- recipients did not express Foxp3 and IL-10 but IFN-γdetermined by FACS. In contract, NICC xenografts in human Treg transferred NOD-SCID IL2rγ-/- recipients still remained intact up to at least 100 days with insulin, glucogan and somatostatin positive staining after rechallenge with CD25+ cell-depleted human PBMC. CD45+ cells which included CD4+ and CD8+ cells didn't infiltrate but surround the graft detected by immunohistology and CD4 positive staining cells and CD4+Foxp3+double positive cells were detectable by immuno-fluorescence double staining. A large number of human CD4+ T cells expressed CD25 and Foxp3 (18.6±2.3%) in human Tregs transferred NICC recipient NOD-SCID IL2ryγ-/- mice at 28 day until to 100 day after rechallenging with human PBMCs. These data suggested that transfer of NOD-SCID IL2rγ-/- recipients with 1 x 107 CD25+ cell-depleted human PBMCs was sufficient to cause NICC xenograft rejection and the human PBMC-mediated xenograft rejection was significantly suppressed by human expanded Tregs which led to a long-term NICC graft survival. High level expression of Treg (Foxp3, CTLA-4, IL-10) but not T effect cell (IFN-y) associated genes was detected in NICC grafts from human Treg transferred NICC recipient mice 28 days after human PBMCs rechallenge by real-time PCR and human Tregs transferred NICC recipients demonstrated reduced level of human IFN-y and elevated level of human IL-10 in their serum 28 days after rechallenge with CD25+ cell-depleted human PBMC detected by cytometric bead array. These data suggest that the prolonged xenograft survival in the human Treg transferred and PBMC rechallenged NOD-SCID IL2ryγ-/- recipients was associated with the presence of human Tregs coexpressing IL-10 and inhibited production of effector cytokine IFN-y in these mice.
     Conclusions:Tissue factor expression in NICC can be reduced by siRNA mediated knockdown and TF knockdown can reduce IBMIR in vitro. Human expanded Tregs were capable of protecting NICC xenograft from rejection mediated by CD25+ cell-depleted human PBMC in NOD-SCID IL2rg-/- recipients and IL-10 was required for human Treg-mediated suppression of the human anti-pig xenogeneic response in vivo. Humanized NOD-SCID IL2rg-/- mice are a suitable mouse model to study human Treg-mediated tolerance and the mechanisms involved in islet xenotransplantation.
引文
[1]Bennet W, Groth CG, Larsson R, et al. Isolated human islets trigger an instant blood mediated inflammatory reaction: implications for intraportal islet transplantation as a treatment for patients with type 1 diabetes. Ups J Med Sci, 2000,105(2):125-133.
    [2]Bennet W, Sundberg B, Lundgren T, et al. Damage to porcine islets of Langerhans after exposure to human blood in vitro, or after intraportal transplantation to cynomologus monkeys:protective effects of sCRl and heparin. Transplantation,2000,69(5):711-719.
    [3]Cantarovich D, Blancho G, Potiron N, et al. Rapid failure of pig islet transplantation in non human primates. Xenotransplantation,2002,9(1):25-35.
    [4]Bennet W, Sundberg B, Groth CG, et al. Incompatibility between human blood and isolated islets of Langerhans:a finding with implications for clinical intraportal islet transplantation? Diabetes,1999,48(10):1907-1914.
    [5]Moberg L. The role of the innate immunity in islet transplantation. Ups J Med Sci,2005,110(1):17-55.
    [6]Ozmen L, Ekdahl KN, Elgue G, et al. Inhibition of thrombin abrogates the instant blood-mediated inflammatory reaction triggered by isolated human islets:possible application of the thrombin inhibitor melagatran in clinical islet transplantation. Diabetes,2002,51(6):1779-1784.
    [7]Cowan PJ, d'Apice AJ. The coagulation barrier in xenotransplantation: incompatibilities and strategies to overcome them. Curr Opin Organ Transplant, 2008,13(2):178-183.
    [8]Crikis S, Cowan PJ, d'Apice AJ. Intravascular thrombosis in discordant xenotransplantation. Transplantation,2006,82(9):1119-1123.
    [9]Cowan PJ. Coagulation and the xenograft endothelium. Xenotransplantation, 2007,14(1):7-12.
    [10]van der Windt DJ, Bottino R, Casu A, et al. Rapid loss of intraportally transplanted islets:an overview of pathophysiology and preventive strategies. Xenotransplantation,2007,14(4):288-297.
    [11]Moberg L, Johansson H, Lukinius A, et al. Production of tissue factor by pancreatic islet cells as a trigger of detrimental thrombotic reactions in clinical islet transplantation. Lancet,2002,360(9350):2039-2045.
    [12]Johansson H, Lukinius A, Moberg L, et al. Tissue factor produced by the endocrine cells of the islets of Langerhans is associated with a negative outcome of clinical islet transplantation. Diabetes,2005,54(6):1755-1762.
    [13]Norris LA. Blood coagulation. Best Pract Res Clin Obstet Gynaecol,2003, 17(3):369-383.
    [14]Berman DM, Cabrera O, Kenyon NM, et al. Interference with tissue factor prolongs intrahepatic islet allograft survival in a nonhuman primate marginal mass model. Transplantation,2007,84(3):308-315.
    [15]Nagata M, Mullen Y, Matsuo S, et al. Destruction of islet isografts by severe nonspecific inflammation. Transplant Proc,1990,22(2):855-856.
    [16]Dorling A, Lombardi G, Binns R, et al. Detection of primary direct and indirect human anti-porcine T cell responses using a porcine dendritic cell population. Eur J Immunol,1996,26(6):1378-1387.
    [17]Olack BJ, Jaramillo A, Benshoff ND, et al. Rejection of porcine islet xenografts mediated by CD4+ T cells activated through the indirect antigen recognition pathway. Xenotransplantation,2002,9(6):393-401.
    [18]Mai G, Bucher P, Morel P, et al. Anti-CD 154 mAb treatment but not recipient CD 154 deficiency leads to long-term survival of xenogeneic islet grafts. Am J Transplant,2005,5(5):1021-1031.
    [19]Koulmanda M, Laufer TM, Auchincloss H, et al. Prolonged survival of fetal pig islet xenografts in mice lacking the capacity for an indirect response. Xenotransplantation,2004, 11(6):525-530.
    [20]Itoh M, Takahashi T, Sakaguchi N, et al. Thymus and autoimmunity: production of CD25+CD4+ naturally anergic and suppressive T cells as a key function of the thymus in maintaining immunologic self-tolerance. J Immunol, 1999,162(9):5317-5326.
    [21]Jordan MS, Boesteanu A, Reed AJ, et al. Thymic selection of CD4+CD25+ regulatory T cells induced by an agonist self-peptide. Nat Immunol,2001, 2(4):301-306.
    [22]Sebille F, Dorling A, Lechler RI. The cellular rejection of xenografts--recent insights. Xenotransplantation,2003,10(1):4-6.
    [23]Hara M, Kingsley CI, Niimi M, et al. IL-10 is required for regulatory T cells to mediate tolerance to alloantigens in vivo. J Immunol,2001,166(6):3789-3796.
    [24]Brouard S, Gagne K, Blancho G, et al. T cell response in xenorecognition and xenografts:a review. Hum Immunol,1999,60(6):455-468.
    [25]Gagliani N, Jofra T, Stabilini A, et al. Antigen-specific dependence of Trl-cell therapy in preclinical models of islet transplant. Diabetes,2010,59(2):433-439.
    [26]Wu J, Yi S, Ouyang L, et al. In vitro expanded human CD4+CD25+ regulatory T cells are potent suppressors of T-cell-mediated xenogeneic responses. Transplantation,2008,85(12):1841-1848.
    [27]Vergani A, D'Addio F, Jurewicz M, et al. A novel clinically relevant strategy to abrogate autoimmunity and regulate alloimmunity in NOD mice. Diabetes, 2010,59(9):2253-2264.
    [28]Zhang D, Sanchez-Fueyo A, Kawamoto K, et al. Thl to Th2 immune deviation facilitates, but does not cause, islet allograft tolerance in mice. Cytokine,2010, 51(3):311-319.
    [29]Emamaullee JA, Stanton L, Schur C, et al. Caspase inhibitor therapy enhances marginal mass islet graft survival and preserves long-term function in islet transplantation. Diabetes,2007,56(5):1289-1298.
    [30]Ito M, Hiramatsu H, Kobayashi K, et al. NOD/SCID/gamma(c)(null) mouse:an excellent recipient mouse model for engraftment of human cells. Blood,2002, 100(9):3175-3182.
    [31]Traggiai E, Chicha L, Mazzucchelli L, et al. Development of a human adaptive immune system in cord blood cell-transplanted mice. Science,2004, 304(5667):104-107.
    [32]Shultz LD, Lyons BL, Burzenski LM, et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells. J Immunol,2005, 174(10):6477-6489.
    [33]Ishikawa F, Yasukawa M, Lyons B, et al. Development of functional human blood and immune systems in NOD/SCID/IL2 receptor{gamma} chain(null) mice. Blood,2005,106(5):1565-1573.
    [34]King M, Pearson T, Shultz LD, et al. A new Hu-PBL model for the study of human islet alloreactivity based on NOD-scid mice bearing a targeted mutation in the IL-2 receptor gamma chain gene. Clin Immunol,2008,126(3):303-314.
    [35]Fire A, Xu S, Montgomery MK, et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature,1998, 391(6669):806-811.
    [36]Korbutt GS, Elliott JF, Ao Z, Smith DK, et al. Large scale isolation, growth, and function of porcine neonatal islet cells. J Clin Invest,1996, 97(9):2119-2129.
    [37]Lin CC, Chen D, McVey JH, et al. Expression of tissue factor and initiation of clotting by human platelets and monocytes after incubation with porcine endothelial cells. Transplantation,2008,86(5):702-709.
    [38]Schwarz DS, Hutvagner G, Haley B, et al. Evidence that siRNAs function as guides, not primers, in the Drosophila and human RNAi pathways. Mol Cell, 2002,10(3):537-548.
    [39]Hannon GJ. RNA interference. Nature,2002,418(6894):244-251.
    [40]Cerutti H. RNA interference:traveling in the cell and gaining functions? Trends Genet,2003,19(1):39-46.
    [41]Lin CC, Cooper DK, Dorling A. Coagulation dysregulation as a barrier to xenotransplantation in the primate. Transpl Immunol,2009,21(2):75-80.
    [42]Gong J, Larsson R, Ekdahl KN, et al. Tubing loops as a model for cardiopulmonary bypass circuits:both the biomaterial and the blood-gas phase interfaces induce complement activation in an in vitro model. J Clin Immunol, 1996,16(4):222-229.
    [43]Farivar RS, Filsoufi F, Adams DH. Mechanisms of Gal(alpha)1-3Gal(beta)1-4GlcNAc-R (alphaGal) expression on porcine valve endothelial cells. J Thorac Cardiovasc Surg,2003,125(2):306-314.
    [44]Hoek JA, Sturk A, ten Cate JW, et al. Laboratory and clinical evaluation of an assay of thrombin-antithrombin III complexes in plasma. Clin Chem,1988, 34(10):2058-2062.
    [45]Ryan EA, Lakey JR, Paty BW, et al. Successful islet transplantation:continued insulin reserve provides long-term glycemic control. Diabetes,2002, 51(7):2148-2157.
    [46]Kirchhof N, Shibata S, Wijkstrom M, et al. Reversal of diabetes in non-immunosuppressed rhesus macaques by intraportal porcine islet xenografts precedes acute cellular rejection. Xenotransplantation,2004, 11(5):396-407.
    [47]van der Windt DJ, Bottino R, Casu A, et al. Long-term controlled normoglycemia in diabetic non-human primates after transplantation with hCD46 transgenic porcine islets. Am J Transplant,2009 Dec, Sect.2716-2726.
    [48]Korsgren O, Nilsson B, Berne C, et al. Current status of clinical islet transplantation. Transplantation,2005,79(10):1289-1293.
    [49]Rood PP, Bottino R, Balamurugan AN, et al. Reduction of early graft loss after intraportal porcine islet transplantation in monkeys. Transplantation,2007, 83(2):202-210.
    [50]Giesen PL, Nemerson Y. Tissue factor on the loose. Semin Thromb Hemost, 2000,26(4):379-384.
    [51]Morrissey JH. Tissue factor: an enzyme cofactor and a true receptor. Thromb Haemost,2001,86(1):66-74.
    [52]Drake TA, Morrissey JH, Edgington TS. Selective cellular expression of tissue factor in human tissues. Implications for disorders of hemostasis and thrombosis. Am J Pathol,1989,134(5):1087-1097.
    [53]Luther T, Flossel C, Mackman N, et al. Tissue factor expression during human and mouse development. Am J Pathol,1996,149(1):101-113.
    [54]Muller I, Klocke A, Alex M, et al. Intravascular tissue factor initiates coagulation via circulating microvesicles and platelets. Faseb J,2003,17(3): 476-478.
    [55]Nemerson Y. Tissue factor:then and now. Thromb Haemost,1995,74(1): 180-184.
    [56]Davie EW, Fujikawa K, Kisiel W. The coagulation cascade:initiation, maintenance, and regulation. Biochemistry,1991,30(43):10363-10370.
    [57]Broze GJ, Jr. Tissue factor pathway inhibitor and the revised theory of coagulation. Annu Rev Med,1995,46:103-112.
    [58]Mackman N. Tissue-specific hemostasis:role of tissue factor. J Thromb Haemost,2008,6(2):303-305.
    [59]Cicala C, Cirino G. Linkage between inflammation and coagulation:an update on the molecular basis of the crosstalk. Life Sci,1998,62(20):1817-1824.
    [60]Moberg L, Olsson A, Berne C, et al. Nicotinamide inhibits tissue factor expression in isolated human pancreatic islets:implications for clinical islet transplantation. Transplantation,2003,76(9):1285-1288.
    [61]Keller TT, Mairuhu AT, de Kruif MD, et al. Infections and endothelial cells. Cardiovasc Res,2003,60(1):40-48.
    [62]Johansson H, Goto M, Dufrane D, et al. Low molecular weight dextran sulfate: a strong candidate drug to block IBMIR in clinical islet transplantation. Am J Transplant,2006,6(2):305-312.
    [63]Cabric S, Sanchez J, Lundgren T, et al. Islet surface heparinization prevents the instant blood-mediated inflammatory reaction in islet transplantation. Diabetes, 2007,56(8):2008-2015.
    [64]Lewis EC, Shapiro L, Bowers OJ, et al. Alpha1-antitrypsin monotherapy prolongs islet allograft survival in mice. Proc Natl Acad Sci U S A,2005, 102(34):12153-12158.
    [65]Pileggi A, Molano RD, Berney T, et al. Heme oxygenase-1 induction in islet cells results in protection from apoptosis and improved in vivo function after transplantation. Diabetes,2001,50(9):1983-1991.
    [66]Campbell SC, Aldibbiat A, Marriott CE, et al. Selenium stimulates pancreatic beta-cell gene expression and enhances islet function. FEBS Lett,2008, 582(15):2333-2337.
    [67]Bottino R, Balamurugan AN, Tse H, et al. Response of human islets to isolation stress and the effect of antioxidant treatment. Diabetes,2004,53(10): 2559-2568.
    [68]Thomas DA, Stauffer C, Zhao K, et al. Mitochondrial targeting with antioxidant peptide SS-31 prevents mitochondrial depolarization, reduces islet cell apoptosis, increases islet cell yield, and improves posttransplantation function. J Am Soc Nephrol,2007,18(1):213-222.
    [69]Eisenbarth GS. Type I diabetes mellitus. A chronic autoimmune disease. N Engl J Med,1986,314(21):1360-1368.
    [70]Atkinson MA, Maclaren NK. The pathogenesis of insulin-dependent diabetes mellitus. N Engl J Med,1994,331(21):1428-1436.
    [71]Shapiro AM, Lakey JR, Ryan EA, et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med,2000,343(4):230-238.
    [72]Mirenda V, Golshayan D, Read J, Berton I, et al. Achieving permanent survival of islet xenografts by independent manipulation of direct and indirect T-cell responses. Diabetes,2005,54(4):1048-1055.
    [73]O'Connell P. Pancreatic islet xenotransplantation. Xenotransplantation,2002, 9(6):367-371.
    [74]Groth CG, Korsgren O, Tibell A, et al. Transplantation of porcine fetal pancreas to diabetic patients. Lancet,1994,344(8934):1402-1404.
    [75]Hering BJ, Wijkstrom M, Graham ML, et al. Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat Med,2006,12(3):301-303.
    [76]Cardona K, Korbutt GS, Milas Z, et al. Long-term survival of neonatal porcine islets in nonhuman primates by targeting costimulation pathways. Nat Med, 2006,12(3):304-306.
    [77]Potiron N, Chagneau C, Boeffard F, et al. Adenovirus-mediated CTLA4Ig or CD40Ig gene transfer delays pancreatic islet rejection in a rat-to-mouse xenotransplantation model after systemic but not local expression. Cell Transplant,2005,14(5):263-275.
    [78]Sun L, Yi S, O'Connell PJ. Foxp3 regulates human natural CD4+CD25+ regulatory T-cell-mediated suppression of xenogeneic response. Xenotransplantation,2010,17(2):121-130.
    [79]Sun L, Yi S, O'Connell PJ. IL-10 is required for human CD4(+)CD25(+) regulatory T cell-mediated suppression of xenogeneic proliferation. Immunol Cell Biol,2010,88(4):477-485.
    [80]Fu Y, Yi S, Wu J, et al. In vitro suppression of xeno immune-mediated macrophage activation by human CD4+CD25+ regulatory T cells. Transplantation,2008,86(6):865-874.
    [81]Flanagan SP.'Nude', a new hairless gene with pleiotropic effects in the mouse. Genet Res,1966,8(3):295-309.
    [82]Bosma GC, Custer RP, Bosma MJ. A severe combined immunodeficiency mutation in the mouse. Nature,1983,301(5900):527-530.
    [83]Mosier DE, Gulizia RJ, Baird SM, et al. Transfer of a functional human immune system to mice with severe combined immunodeficiency. Nature,1988, 335(6187):256-259.
    [84]McCune JM, Namikawa R, Kaneshima H, et al. The SCID-hu mouse:murine model for the analysis of human hematolymphoid differentiation and function. Science,1988,241(4873):1632-1639.
    [85]Shultz LD, Schweitzer PA, Christianson SW, et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol,1995, 154(1):180-191.
    [86]Hesselton RM, Greiner DL, Mordes JP, et al. High levels of human peripheral blood mononuclear cell engraftment and enhanced susceptibility to human immunodeficiency virus type 1 infection in NOD/LtSz-scid/scid mice. J Infect Dis,1995,172(4):974-982.
    [87]Lowry PA, Shultz LD, Greiner DL, et al. Improved engraftment of human cord blood stem cells in NOD/LtSz-scid/scid mice after irradiation or multiple-day injections into unirradiated recipients. Biol Blood Marrow Transplant,1996, 2(1):15-23.
    [88]Cao X, Shores EW, Hu-Li J, et al. Defective lymphoid development in mice lacking expression of the common cytokine receptor gamma chain. Immunity, 1995,2(3):223-238.
    [89]DiSanto JP, Muller W, Guy-Grand D, et al. Lymphoid development in mice with a targeted deletion of the interleukin 2 receptor gamma chain. Proc Natl Acad Sci U S A,1995,92(2):377-381.
    [90]Ohbo K, Suda T, Hashiyama M, et al. Modulation of hematopoiesis in mice with a truncated mutant of the interleukin-2 receptor gamma chain. Blood, 1996,87(3):956-967.
    [91]Piccirillo CA, Letterio JJ, Thornton AM, et al. CD4(+)CD25(+) regulatory T cells can mediate suppressor function in the absence of transforming growth factor betal production and responsiveness. J Exp Med,2002,196(2):237-246.
    [92]Takahashi T, Kuniyasu Y, Toda M, et al. Immunologic self-tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease by breaking their anergic/suppressive state. Int Immunol,1998,10(12):1969-1980.
    [93]Suri-Payer E, Cantor H. Differential cytokine requirements for regulation of autoimmune gastritis and colitis by CD4(+)CD25(+) T cells. J Autoimmun, 2001,16(2):115-123.
    [94]Josien R, Douillard P, Guillot C, et al. A critical role for transforming growth factor-beta in donor transfusion-induced allograft tolerance. J Clin Invest,1998, 102(11):1920-1926.
    [95]Blom L, Nystrom L, Dahlquist G. The Swedish childhood diabetes study. Vaccinations and infections as risk determinants for diabetes in childhood. Diabetologia,1991,34(3):176-181.
    [96]Dahlquist G, Mustonen L. Childhood onset diabetes--time trends and climatological factors. Int J Epidemiol,1994,23(6):1234-1241.
    [97]King H, Aubert RE, Herman WH. Global burden of diabetes,1995-2025: prevalence, numerical estimates, and projections. Diabetes Care,1998, 21(9):1414-1431.
    [98]Amos AF, McCarty DJ, Zimmet P. The rising global burden of diabetes and its complications:estimates and projections to the year 2010. Diabet Med,1997, 14 Suppl 5:S1-85.
    [99]Robertson RP, Davis C, Larsen J, et al. Pancreas and islet transplantation for patients with diabetes. Diabetes Care,2000,23(1):112-116.
    [100]Menger MD, Yamauchi J, Vollmar B. Revascularization and microcirculation of freely grafted islets of Langerhans. World J Surg,2001,25(4):509-515.
    [101]Bretzel RG, Hopt UT, Schatz H. Transplantations in patients with diabetes mellitus. Exp Clin Endocrinol Diabetes,2000,108(4):241-242.
    [102]Davey MG, Luscher EF. Actions of thrombin and other coagulant and proteolytic enzymes on blood platelets. Nature,1967,216(5118):857-858.
    [103]Henn V, Slupsky JR, Grafe M, et al. CD40 ligand on activated platelets triggers an inflammatory reaction of endothelial cells. Nature,1998,391(6667): 591-594.
    [104]Stenberg PE, McEver RP, Shuman MA, et al. A platelet alpha-granule membrane protein (GMP-140) is expressed on the plasma membrane after activation. J Cell Biol,1985,101(3):880-886.
    [105]Hughes PE, Pfaff M. Integrin affinity modulation. Trends Cell Biol,1998,8(9): 359-364.
    [106]Krishnaswamy S, Field KA, Edgington TS, et al. Role of the membrane surface in the activation of human coagulation factor Ⅹ. J Biol Chem,1992,267(36): 26110-26120.
    [107]Werling RW, Zacharski LR, Kisiel W, et al. Distribution of tissue factor pathway inhibitor in normal and malignant human tissues. Thromb Haemost, 1993,69(4):366-369.
    [108]Bajaj MS, Kuppuswamy MN, Saito H, et al. Cultured normal human hepatocytes do not synthesize lipoprotein-associated coagulation inhibitor: evidence that endothelium is the principal site of its synthesis. Proc Natl Acad Sci U S A,1990,87(22):8869-8873.
    [109]Sandset PM. Tissue factor pathway inhibitor (TFPI)--an update. Haemostasis, 1996,26 Suppl 4:154-165.
    [110]Novotny WF, Girard TJ, Miletich JP, et al. Purification and characterization of the lipoprotein-associated coagulation inhibitor from human plasma. J Biol Chem,1989,264(31):18832-18837.
    [111]Ye J, Esmon NL, Esmon CT, et al. The active site of thrombin is altered upon binding to thrombomodulin. Two distinct structural changes are detected by fluorescence, but only one correlates with protein C activation. J Biol Chem, 1991,266(34):23016-23021.
    [112]Esmon NL, Owen WG, Esmon CT. Isolation of a membrane-bound cofactor for thrombin-catalyzed activation of protein C. J Biol Chem,1982,257(2): 859-864.
    [113]Bajzar L, Manuel R, Nesheim ME. Purification and characterization of TAFI, a thrombin-activable fibrinolysis inhibitor. J Biol Chem,1995,270(24): 14477-14484.
    [114]Camiolo SM, Thorsen S, Astrup T. Fibrinogenolysis and fibrinolysis with tissue plasminogen activator, urokinase, streptokinase-activated human globulin, and plasmin. Proc Soc Exp Biol Med,1971,138(1):277-280.
    [115]Conkling PR, Greenberg CS, Weinberg JB. Tumor necrosis factor induces tissue factor-like activity in human leukemia cell line U937 and peripheral blood monocytes. Blood,1988,72(1):128-133.
    [116]Froberg MK, Leone JP, Jessurun J, et al. Fatal disseminated intravascular coagulation after autologous islet transplantation. Hum Pathol,1997, 28(11):1295-1298.
    [117]Shapiro AM, Lakey JR, Rajotte RV, et al. Portal vein thrombosis after transplantation of partially purified pancreatic islets in a combined human liver/islet allograft. Transplantation,1995,59(7):1060-1063.
    [118]Gustafsson D, Antonsson T, Bylund R, et al. Effects of melagatran, a new low-molecular-weight thrombin inhibitor, on thrombin and fibrinolytic enzymes. Thromb Haemost,1998,79(1):110-118.
    [119]Mehta JL, Chen L, Nichols WW, et al. Melagatran, an oral active-site inhibitor of thrombin, prevents or delays formation of electrically induced occlusive thrombus in the canine coronary artery. J Cardiovasc Pharmacol,1998,31(3): 345-351.
    [120]Moberg L, Korsgren O, Nilsson B. Neutrophilic granulocytes are the predominant cell type infiltrating pancreatic islets in contact with ABO-compatible blood. Clin Exp Immunol,2005,142(1):125-131.
    [121]Tanoue M, Yoshizawa Y, Sato T, Yano H, et al. The role of complement-derived chemotactic factors in lung injury induced by preformed immune complexes. Int Arch Allergy Immunol,1993,101(1):47-51.
    [122]Piemonti L, Leone BE, Nano R, et al. Human pancreatic islets produce and secrete MCP-1/CCL2:relevance in human islet transplantation. Diabetes,2002, 51(1):55-65.
    [123]Johansson U, Olsson A, Gabrielsson S, et al. Inflammatory mediators expressed in human islets of Langerhans:implications for islet transplantation. Biochem Biophys Res Commun,2003,308(3):474-479.
    [124]Cattan P, Berney T, Schena S, et al. Early assessment of apoptosis in isolated islets of Langerhans. Transplantation,2001,71(7):857-862.
    [125]Contreras JL, Jenkins S, Eckhoff DE, et al. Stable alpha-and beta-islet cell function after tolerance induction to pancreatic islet allografts in diabetic primates. Am J Transplant,2003,3(2):128-138.
    [126]Emamaullee JA, Shapiro AM. Interventional strategies to prevent beta-cell apoptosis in islet transplantation. Diabetes,2006,55(7):1907-1914.
    [127]Villiger P, Ryan EA, Owen R, et al. Prevention of bleeding after islet transplantation:lessons learned from a multivariate analysis of 132 cases at a single institution. Am J Transplant,2005,5(12):2992-2998.
    [128]Goto M, Johansson H, Maeda A, et al. Low molecular weight dextran sulfate prevents the instant blood-mediated inflammatory reaction induced by adult porcine islets. Transplantation,2004,77(5):741-747.
    [129]Cui W, Wilson JT, Wen J, et al. Thrombomodulin improves early outcomes after intraportal islet transplantation. Am J Transplant,2009,9(6):1308-1316.
    [130]Beuneu C, Vosters O, Ling Z, et al. N-Acetylcysteine derivative inhibits procoagulant activity of human islet cells. Diabetologia,2007,50(2):343-347.
    [131]Arita S, Nagai T, Ochiai M, et al. Prevention of primary nonfunction of canine islet autografts by treatment with pravastatin. Transplantation,2002, 73(1):7-12.
    [132]Orvim U, Barstad RM, Orning L, et al. Antithrombotic efficacy of inactivated active site recombinant factor Ⅶa is shear dependent in human blood. Arterioscler Thromb Vase Biol,1997,17(11):3049-3056.
    [133]Soderstrom T, Hedner U, Arnljots B. Active site-inactivated factor Ⅶa prevents thrombosis without increased surgical bleeding:topical and intravenous administration in a rat model of deep arterial injury. J Vase Surg, 2001,33(5):1072-1079.
    [134]Arnljots B, Ezban M, Hedner U. Prevention of experimental arterial thrombosis by topical administration of active site-inactivated factor Ⅶa. J Vase Surg, 1997,25(2):341-346.
    [135]Golino P, Ragni M, Cirillo P, et al. Antithrombotic effects of recombinant human, active site-blocked factor Ⅶa in a rabbit model of recurrent arterial thrombosis. Circ Res,1998,82(1):39-46.
    [136]Lev El, Marmur JD, Zdravkovic M, et al. Antithrombotic effect of tissue factor inhibition by inactivated factor Ⅶa:an ex vivo human study. Arterioscler Thromb Vasc Biol,2002,22(6):1036-1041.
    [137]Erhardtsen E, Nilsson P, Johannessen M, et al. Pharmacokinetics and safety of FFR-rFVIIa after single doses in healthy subjects. J Clin Pharmacol,2001, 41(8):880-885.
    [138]Mackman N, Brand K, Edgington TS. Lipopolysaccharide-mediated transcriptional activation of the human tissue factor gene in THP-1 monocytic cells requires both activator protein 1 and nuclear factor kappa B binding sites. J Exp Med,1991,174(6):1517-1526.
    [139]Ueda A, Okuda K, Ohno S, et al. NF-kappa B and Spl regulate transcription of the human monocyte chemoattractant protein-1 gene. J Immunol,1994, 153(5):2052-2063.
    [140]Korsgren O, Andersson A, Sandlcr S. Pretreatment of fetal porcine pancreas in culture with nicotinamide accelerates reversal of diabetes after transplantation to nude mice. Surgery,1993,113(2):205-214.
    [141]Kolb H, Burkart V, Appels B, et al. Essential contribution of macrophages to islet cell destruction in vivo and in vitro. J Autoimmun,1990,3 Suppl 1:117-120.
    [142]Buscema M, Vinci C, Gatta C, et al. Nicotinamide partially reverses the interleukin-1 beta inhibition of glucose-induced insulin release in pancreatic islets. Metabolism,1992,41(3):296-300.
    [143]Kallmann B, Burkart V, Kroncke KD, et al. Toxicity of chemically generated nitric oxide towards pancreatic islet cells can be prevented by nicotinamide. Life Sci,1992,51(9):671-678.
    [144]Bottino R, Balamurugan AN, Bertera S, et al. Preservation of human islet cell functional mass by anti-oxidative action of a novel SOD mimic compound. Diabetes,2002,51(8):2561-2567.
    [145]Wang H, Lee SS, Dell'Agnello C, et al. Bilirubin can induce tolerance to islet allografts. Endocrinology,2006,147(2):762-768.
    [146]Wang H, Lee SS, Gao W, et al. Donor treatment with carbon monoxide can yield islet allograft survival and tolerance. Diabetes,2005,54(5):1400-1406.
    [147]Xie D, Smyth CA, Eckstein C, et al. Cytoprotection of PEG-modified adult porcine pancreatic islets for improved xenotransplantation. Biomaterials,2005, 26(4):403-412.
    [148]Miura S, Teramura Y, Iwata H. Encapsulation of islets with ultra-thin polyion complex membrane through poly(ethylene glycol)-phospholipids anchored to cell membrane. Biomaterials,2006,27(34):5828-5835.
    [149]Kim HI, Yu JE, Lee SY, et al. The effect of composite pig islet-human endothelial cell grafts on the instant blood-mediated inflammatory reaction. Cell Transplant,2009,18(1):31-37.
    [150]Emamaullee JA, Rajotte RV, Liston P, et al. XIAP overexpression in human islets prevents early posttransplant apoptosis and reduces the islet mass needed to treat diabetes. Diabetes,2005,54(9):2541-2548.
    [151]Mysore TB, Shinkel TA, Collins J, et al. Overexpression of glutathione peroxidase with two isoforms of superoxide dismutase protects mouse islets from oxidative injury and improves islet graft function. Diabetes,2005, 54(7):2109-2116.
    [152]Moriscot C, Pattou F, Kerr-Conte J, et al. Contribution of adenoviral-mediated superoxide dismutase gene transfer to the reduction in nitric oxide-induced cytotoxicity on human islets and INS-1 insulin-secreting cells. Diabetologia, 2000,43(5):625-631.
    [153]Dwyer KM, Mysore TB, Crikis S, et al. The transgenic expression of human CD39 on murine islets inhibits clotting of human blood. Transplantation,2006, 82(3):428-432.
    [154]Schmidt P, Goto M, Le Mauff B, et al. Adenovirus-mediated expression of human CD55 or CD59 protects adult porcine islets from complement-mediated cell lysis by human serum. Transplantation,2003,75(5):697-702.
    [155]Dunn IF, Heese O, Black PM. Growth factors in glioma angiogenesis:FGFs, PDGF, EGF, and TGFs. J Neurooncol,2000,50(1-2):121-137.
    [156]Jensen RL. Growth factor-mediated angiogenesis in the malignant progression of glial tumors:a review. Surg Neurol,1998,49(2):189-195; discussion 196.