共查询到20条相似文献,搜索用时 36 毫秒
1.
Prevention of Allograft Rejection by Use of Regulatory T Cells With an MHC‐Specific Chimeric Antigen Receptor 下载免费PDF全文
F. Noyan K. Zimmermann M. Hardtke‐Wolenski A. Knoefel E. Schulde R. Geffers M. Hust J. Huehn M. Galla M. Morgan A. Jokuszies M. P. Manns E. Jaeckel 《American journal of transplantation》2017,17(4):917-930
CD4+CD25highFOXP3+ regulatory T cells (Tregs) are involved in graft‐specific tolerance after solid organ transplantation. However, adoptive transfer of polyspecific Tregs alone is insufficient to prevent graft rejection even in rodent models, indicating that graft‐specific Tregs are required. We developed a highly specific chimeric antigen receptor that recognizes the HLA molecule A*02 (referred to as A2‐CAR). Transduction into natural regulatory T cells (nTregs) changes the specificity of the nTregs without alteration of their regulatory phenotype and epigenetic stability. Activation of nTregs via the A2‐CAR induced proliferation and enhanced the suppressor function of modified nTregs. Compared with nTregs, A2‐CAR Tregs exhibited superior control of strong allospecific immune responses in vitro and in humanized mouse models. A2‐CAR Tregs completely prevented rejection of allogeneic target cells and tissues in immune reconstituted humanized mice in the absence of any immunosuppression. Therefore, these modified cells have great potential for incorporation into clinical trials of Treg‐supported weaning after allogeneic transplantation. 相似文献
2.
Aquaporin 4 blockade improves survival of murine heart allografts subjected to prolonged cold ischemia 下载免费PDF全文
Katayoun Ayasoufi Naoki Kohei Michael Nicosia Ran Fan George W. Farr Paul R. McGuirk Marc F. Pelletier Robert L. Fairchild Anna Valujskikh 《American journal of transplantation》2018,18(5):1238-1246
Prolonged cold ischemia storage (CIS) is a leading risk factor for poor transplant outcome. Existing strategies strive to minimize ischemia–reperfusion injury in transplanted organs, yet there is a need for novel approaches to improve outcomes of marginal allografts and expand the pool of donor organs suitable for transplantation. Aquaporins (AQPs) are a family of water channels that facilitate homeostasis, tissue injury, and inflammation. We tested whether inhibition of AQP4 improves the survival of fully MHC‐mismatched murine cardiac allografts subjected to 8 hours of CIS. Administration of a small molecule AQP4 inhibitor during donor heart collection and storage and for a short‐time posttransplantation improves the viability of donor graft cells, diminishes donor‐reactive T cell responses, and extends allograft survival in the absence of other immunosuppression. Furthermore, AQP4 inhibition is synergistic with cytotoxic T lymphocyte–associated antigen 4–Ig in prolonging survival of 8‐hour CIS heart allografts. AQP4 blockade markedly reduced T cell proliferation and cytokine production in vitro, suggesting that the improved graft survival is at least in part mediated through direct effects on donor‐reactive T cells. These results identify AQPs as a promising target for diminishing donor‐specific alloreactivity and improving the survival of high‐risk organ transplants. 相似文献
3.
In Vitro Induction of Human Regulatory T Cells Using Conditions of Low Tryptophan Plus Kynurenines 下载免费PDF全文
K. L. Hippen R. S. O'Connor A. M. Lemire A. Saha E. A. Hanse N. C. Tennis S. C. Merkel A. Kelekar J. L. Riley B. L. Levine C. H. June L. A. Turka L. S. Kean M. L. MacMillan J. S. Miller J. E. Wagner D. H. Munn B. R. Blazar 《American journal of transplantation》2017,17(12):3098-3113
Thymic regulatory T cells (tTregs) and induced regulatory T cells (iTregs) suppress murine acute graft‐versus‐host disease (GVHD). Previously, we demonstrated that the plasmacytoid dendritic cell indoleamine 2,3‐dioxygenase (IDO) fosters the in vitro development of human iTregs via tryptophan depletion and kynurenine (Kyn) metabolites. We now show that stimulation of naïve CD4+ T cells in low tryptophan (low Trp) plus Kyn supports human iTreg generation. In vitro, low Trp + Kyn iTregs and tTregs potently suppress T effector cell proliferation equivalently but are phenotypically distinct. Compared with tTregs or T effector cells, bioenergetics profiling reveals that low Trp + Kyn iTregs have increased basal glycolysis and oxidative phosphorylation and use glutaminolysis as an energy source. Low Trp + Kyn iTreg viability was reliant on interleukin (IL)‐2 in vitro. Although in vivo IL‐2 administration increased low Trp + Kyn iTreg persistence on adoptive transfer into immunodeficient mice given peripheral blood mononuclear cells to induce GVHD, IL‐2–supported iTregs did not improve recipient survival. We conclude that low Trp + Kyn create suppressive iTregs that have high metabolic needs that will need to be addressed before clinical translation. 相似文献
4.
Giulia Coco William Foulsham Takeshi Nakao Jia Yin Afsaneh Amouzegar Yukako Taketani Sunil K. Chauhan Reza Dana 《American journal of transplantation》2020,20(2):389-398
The functional competence of corneal endothelial cells (CEnCs) is critical for survival of corneal allografts, but these cells are often targets of the immune response mediated by graft‐attacking effector T cells. Although regulatory T cells (Tregs) have been studied for their role in regulating the host’s alloimmune response towards the graft, the cytoprotective function of these cells on CEnCs has not been investigated. The aim of this study was to determine whether Tregs suppress effector T cell–mediated and inflammatory cytokine–induced CEnC death, and to elucidate the mechanism by which this cytoprotection occurs. Using 2 well‐established models of corneal transplantation (low‐risk and high‐risk models), we show that Tregs derived from low‐risk graft recipients have a superior capacity in protecting CEnCs against effector T cell–mediated and interferon‐γ and tumor necrosis factor‐α‐induced cell death compared to Tregs derived from high‐risk hosts. We further demonstrate that the cytoprotective function of Tregs derived from low‐risk hosts occurs independently of direct cell‐cell contact and is mediated by the immunoregulatory cytokine IL‐10. Our study is the first to report that Tregs provide cytoprotection for CEnCs through secretion of IL‐10, indicating potentially novel therapeutic targets for enhancing CEnC survival following corneal transplantation. 相似文献
5.
Adoptive Transfer of Tracer‐Alloreactive CD4+ T Cell Receptor Transgenic T Cells Alters the Endogenous Immune Response to an Allograft 下载免费PDF全文
M. L. Miller J. Chen M. D. Daniels M. G. McKeague Y. Wang D. Yin V. Vu M.‐L. Alegre 《American journal of transplantation》2016,16(10):2842-2853
T cell receptor transgenic (TCR‐Tg) T cells are often used as tracer populations of antigen‐specific responses to extrapolate findings to endogenous T cells. The extent to which TCR‐Tg T cells behave purely as tracer cells or modify the endogenous immune response is not clear. To test the impact of TCR‐Tg T cell transfer on endogenous alloimmunity, recipient mice were seeded with CD4+ or CD8+ TCR‐Tg or polyclonal T cells at the time of cardiac allograft transplantation. Only CD4+ TCR‐Tg T cells accelerated rejection and, unexpectedly, led to a dose‐dependent decrease in both transferred and endogenous T cells infiltrating the graft. In contrast, recipients of CD4+ TCR‐Tg T cells exhibited enhanced endogenous donor‐specific CD8+ T cell activation in the spleen and accelerated alloantibody production. Introduction of CD4+ TCR‐Tg T cells also perturbed the intragraft accumulation of innate cell populations. Transferred CD4+ TCR‐Tg T cells alter many aspects of endogenous alloimmunity, suggesting that caution should be used when interpreting experiments using these adoptively transferred cells because the overall nature of allograft rejection may be altered. These results also may have implications for adoptive CD4+ T cell immunotherapy in tumor and infectious clinical settings because cell infusion may have additional effects on natural immune responses. 相似文献
6.
Complement Dependence of Murine Costimulatory Blockade‐Resistant Cellular Cardiac Allograft Rejection 下载免费PDF全文
N. Chun R. L. Fairchild Y. Li J. Liu M. Zhang W. M. Baldwin III P. S. Heeger 《American journal of transplantation》2017,17(11):2810-2819
Building on studies showing that ischemia–reperfusion‐(I/R)‐injury is complement dependent, we tested links among complement activation, transplantation‐associated I/R injury, and murine cardiac allograft rejection. We transplanted BALB/c hearts subjected to 8‐h cold ischemic storage (CIS) into cytotoxic T‐lymphocyte associated protein 4 (CTLA4)Ig‐treated wild‐type (WT) or c3?/? B6 recipients. Whereas allografts subjected to 8‐h CIS rejected in WT recipients with a median survival time (MST) of 37 days, identically treated hearts survived >60 days in c3?/? mice (p < 0.05, n = 4–6/group). Mechanistic studies showed recipient C3 deficiency prevented induction of intragraft and serum chemokines/cytokines and blunted the priming, expansion, and graft infiltration of interferon‐γ–producing, donor‐reactive T cells. MST of hearts subjected to 8‐h CIS was >60 days in mannose binding lectin (mbl1?/?mbl2?/?) recipients and 42 days in factor B (cfb?/?) recipients (n = 4–6/group, p < 0.05, mbl1?/?mbl2?/? vs. cfb?/?), implicating the MBL (not alternative) pathway. To pharmacologically target MBL‐initiated complement activation, we transplanted BALB/c hearts subjected to 8‐h CIS into CTLA4Ig‐treated WT B6 recipients with or without C1 inhibitor (C1‐INH). Remarkably, peritransplantation administration of C1‐INH prolonged graft survival (MST >60 days, p < 0.05 vs. controls, n = 6) and prevented CI‐induced increases in donor‐reactive, IFNγ‐producing spleen cells (p < 0.05). These new findings link donor I/R injury to T cell–mediated rejection through MBL‐initiated, complement activation and support testing C1‐INH administration to prevent CTLA4Ig‐resistant rejection of deceased donor allografts in human transplant patients. 相似文献
7.
Antoine Sicard Megan K. Levings David W. Scott 《American journal of transplantation》2018,18(6):1305-1311
Adoptive cell therapy with therapeutic T cells has become one of the most promising strategies to stimulate or suppress immune responses. Using virus‐mediated genetic manipulation, the antigen specificity of T cells can now be precisely redirected. Tailored specificity has not only overcome technical limitations and safety concerns but also considerably broadened the spectrum of therapeutic applications. Different T cell–engineering strategies have now become available to suppress alloimmune responses. We first provide an overview of the allorecognition pathways and effector mechanisms that are responsible for alloimmune injuries in the setting of vascularized organ transplantation. We then discuss the potential to use different T cell–engineering approaches to suppress alloimmune responses. Specifically, expression of allospecific T cell receptors, single‐chain chimeric antigen receptors, or antigen domains recognized by B cell receptors (B cell antibody receptors) in regulatory or cytotoxic T cells are considered. The ability of these strategies to control the direct or indirect pathways of allorecognition and the cellular or humoral alloimmune responses is discussed. An intimate understanding of the complex interplay that occurs between the engineered T cells and the alloimmune players is a necessary prerequisite for the design of safe and successful strategies for precise immunomodulation in transplantation. 相似文献
8.
Transplant rejection mediated by the adaptive immune system remains a major barrier to achieving long‐term tolerance and graft survival. Emerging evidence indicates that lymphocytes rapidly shift their metabolic programs in response to activation, co‐stimulatory, and cytokine signals to support required effector cell differentiation and function. These observations have led to the hypothesis that manipulating the metabolic programs of immune cells could serve as a powerful therapeutic strategy for attenuating deleterious immune responses and facilitating durable tolerance in the setting of allogeneic solid organ or bone marrow transplant. In this mini‐review, we introduce the fundamentals of metabolism, highlight the current understanding of how adaptive immune cells utilize their metabolic programs, and discuss the potential for targeting metabolism as a therapeutic approach to induce tolerance in the transplant setting. 相似文献
9.
Therapies using thymus‐derived regulatory T cells (Tregs) are promising strategies for preventing autoimmunity or graft rejection. The efficacy of these approaches is, however, contingent on a better understanding of Treg mode of action, especially about factors controlling their activation in vivo. Although key parameters of Treg suppression have been identified, little information is available on Treg activation in vivo via the TCR. In light of recent studies using TCR transgenic mouse models as well as unpublished data, we discuss evidence in support of the view that Treg TCR specificities are not necessarily highly diverse, that the accessibility of Treg selective antigens control Treg development, and that peptides derived from MHC class II (MHC‐II) could be prevailing antigens involved in Treg selection. This novel perspective provides insights on Treg development as well as a conceptual basis to a significant contribution of MHC‐II derived peptides in the shaping of the Treg TCR repertoire. 相似文献
10.
Regulatory T Cell Infusion Can Enhance Memory T Cell and Alloantibody Responses in Lymphodepleted Nonhuman Primate Heart Allograft Recipients 下载免费PDF全文
M. B. Ezzelarab H. Zhang H. Guo L. Lu A. F. Zahorchak R. W. Wiseman M. A. Nalesnik J. K. Bhama D. K. C. Cooper A. W. Thomson 《American journal of transplantation》2016,16(7):1999-2015
The ability of regulatory T cells (Treg) to prolong allograft survival and promote transplant tolerance in lymphodepleted rodents is well established. Few studies, however, have addressed the therapeutic potential of adoptively transferred, CD4+CD25+CD127?Foxp3+ (Treg) in clinically relevant large animal models. We infused ex vivo–expanded, functionally stable, nonselected Treg (up to a maximum cumulative dose of 1.87 billion cells) into antithymocyte globulin–lymphodepleted, MHC‐mismatched cynomolgus monkey heart graft recipients before homeostatic recovery of effector T cells. The monkeys also received tacrolimus, anti–interleukin‐6 receptor monoclonal antibodies and tapered rapamycin maintenance therapy. Treg administration in single or multiple doses during the early postsurgical period (up to 1 month posttransplantation), when host T cells were profoundly depleted, resulted in inferior graft function compared with controls. This was accompanied by increased incidences of effector memory T cells, enhanced interferon‐γ production by host CD8+ T cells, elevated levels of proinflammatory cytokines, and antidonor alloantibodies. The findings caution against infusion of Treg during the early posttransplantation period after lymphodepletion. Despite marked but transient increases in Treg relative to endogenous effector T cells and use of reputed “Treg‐friendly” agents, the host environment/immune effector mechanisms instigated under these conditions can perturb rather than favor the potential therapeutic efficacy of adoptively transferred Treg. 相似文献
11.
Kaempferol Promotes Transplant Tolerance by Sustaining CD4+FoxP3+ Regulatory T Cells in the Presence of Calcineurin Inhibitor 下载免费PDF全文
Q. Xie S. M. Xu X. W. Jin W. Li A. Zhou Z. Dai 《American journal of transplantation》2015,15(7):1782-1792
Calcineurin inhibitor cyclosporine is widely used as an immunosuppressant in clinic. However, mounting evidence has shown that cyclosporine hinders tolerance induction by dampening Tregs. Therefore, it is of paramount importance to overcome this pitfall. Kaempferol was reported to inhibit DC function. Here, we found that kaempferol delayed islet allograft rejection. Combination of kaempferol and low‐dose, but not high‐dose, of cyclosporine induced allograft tolerance in majority of recipient mice. Although kaempferol plus either dose of cyclosporine largely abrogated proliferation of graft‐infiltrating T cells and their CTL activity, both proliferation and CTL activity in mice treated with kaempferol plus low‐dose, but not high‐dose, cyclosporine reemerged rapidly upon treatment withdrawal. Kaempferol increased CD4+FoxP3+ Tregs both in transplanted mice and in vitro, likely by suppressing DC maturation and their IL‐6 expression. Reduction in Tregs by low dose of cyclosporine was reversed by kaempferol. Kaempferol‐induced Tregs exhibited both allospecific and non‐allospecific suppression. Administering IL‐6 abrogated allograft tolerance induced by kaempferol and cyclosporine via diminishing CD4+FoxP3+ Tregs. Thus, for the first time, we demonstrated that kaempferol promotes transplant tolerance in the presence of low dose of cyclosporine, which allows for sufficient Treg generation while minimizing side effects, resulting in much‐needed synergy between kaempferol and cyclosporine. 相似文献
12.
Control of Immune Response to Allogeneic Embryonic Stem Cells by CD3 Antibody–Mediated Operational Tolerance Induction 下载免费PDF全文
D. Calderon M. Prot S. You C. Marquet V. Bellamy P. Bruneval F. Valette P. de Almeida J. C. Wu M. Pucéat P. Menasché L. Chatenoud 《American journal of transplantation》2016,16(2):454-467
Implantation of embryonic stem cells (ESCs) and their differentiated derivatives into allogeneic hosts triggers an immune response that represents a hurdle to clinical application. We established in autoimmunity and in transplantation that CD3 antibody therapy induces a state of immune tolerance. Promising results have been obtained with CD3 antibodies in the clinic. In this study, we tested whether this strategy can prolong the survival of undifferentiated ESCs and their differentiated derivatives in histoincompatible hosts. Recipients of either mouse ESC–derived embryoid bodies (EBs) or cardiac progenitors received a single short tolerogenic regimen of CD3 antibody. In immunocompetent mice, allogeneic EBs and cardiac progenitors were rejected within 20–25 days. Recipients treated with CD3 antibody showed long‐term survival of implanted cardiac progenitors or EBs. In due course, EBs became teratomas, the growth of which was self‐limited. Regulatory CD4+FoxP3+ T cells and signaling through the PD1/PDL1 pathway played key roles in the CD3 antibody therapeutic effect. Gene profiling emphasized the importance of TGF‐β and the inhibitory T cell coreceptor Tim3 to the observed effect. These results demonstrate that CD3 antibody administered alone promotes prolonged survival of allogeneic ESC derivatives and thus could prove useful for enhancing cell engraftment in the absence of chronic immunosuppression. 相似文献
13.
In Vivo Mobilization and Functional Characterization of Nonhuman Primate Monocytic Myeloid‐Derived Suppressor Cells 下载免费PDF全文
A. F. Zahorchak M. B. Ezzelarab L. Lu H. R. Turnquist A. W. Thomson 《American journal of transplantation》2016,16(2):661-671
14.
Mostafa T. Mohammed Songjie Cai Benjamin L. Hanson Hengcheng Zhang Rachel L. Clement Joe Daccache Cecilia B. Cavazzoni Bruce R. Blazar Alessandro Alessandrini Helmut G. Rennke Anil Chandraker Peter T. Sage 《American journal of transplantation》2021,21(5):1893-1901
Following solid organ transplantation, a substantial proportion of chronic allograft loss is attributed to the formation of donor-specific antibodies (DSAs) and antibody-mediated rejection (AbMR). The frequency and phenotype of T follicular helper (Tfh) and T follicular regulatory (Tfr) cells is altered in the setting of kidney transplantation, particularly in patients who develop AbMR. However, the roles of Tfh and Tfr cells in AbMR after solid organ transplantation is unclear. We developed mouse models to inducibly and potently perturb Tfh and Tfr cells to assess the roles of these cells in the development of DSA and AbMR. We found that Tfh cells are required for both de novo DSA responses as well as augmentation of DSA following presensitization. Using orthotopic allogeneic kidney transplantation models, we found that deletion of Tfh cells at the time of transplantation resulted in less severe transplant rejection. Furthermore, using inducible Tfr cell deletion strategies we found that Tfr cells inhibit de novo DSA formation but only have a minor role in controlling kidney transplant rejection. These studies demonstrate that Tfh cells promote, whereas Tfr cells inhibit, DSA to control rejection after kidney transplantation. Therefore, targeting these cells represent a new therapeutic strategy to prevent and treat AbMR. 相似文献
15.
Sequential Monitoring and Stability of Ex Vivo–Expanded Autologous and Nonautologous Regulatory T Cells Following Infusion in Nonhuman Primates 下载免费PDF全文
H. Zhang H. Guo L. Lu A. F. Zahorchak R. W. Wiseman G. Raimondi D. K. C. Cooper M. B. Ezzelarab A. W. Thomson 《American journal of transplantation》2015,15(5):1253-1266
16.
Renal Allograft Survival in Nonhuman Primates Infused With Donor Antigen‐Pulsed Autologous Regulatory Dendritic Cells 下载免费PDF全文
M. B. Ezzelarab D. Raich‐Regue L. Lu A. F. Zahorchak A. Perez‐Gutierrez A. Humar M. Wijkstrom M. Minervini R. W. Wiseman D. K. C. Cooper A. E. Morelli A. W. Thomson 《American journal of transplantation》2017,17(6):1476-1489
Systemic administration of autologous regulatory dendritic cells (DCreg; unpulsed or pulsed with donor antigen [Ag]), prolongs allograft survival and promotes transplant tolerance in rodents. Here, we demonstrate that nonhuman primate (NHP) monocyte‐derived DCreg preloaded with cell membrane vesicles from allogeneic peripheral blood mononuclear cells induce T cell hyporesponsiveness to donor alloantigen (alloAg) in vitro. These donor alloAg‐pulsed autologous DCreg (1.4–3.6 × 106/kg) were administered intravenously, 1 day before MHC‐mismatched renal transplantation to rhesus monkeys treated with costimulation blockade (cytotoxic T lymphocyte Ag 4 immunoglobulin [CTLA4] Ig) and tapered rapamycin. Prolongation of graft median survival time from 39.5 days (no DCreg infusion; n = 6 historical controls) and 29 days with control unpulsed DCreg (n = 2), to 56 days with donor Ag‐pulsed DCreg (n = 5) was associated with evidence of modulated host CD4+ and CD8+ T cell responses to donor Ag and attenuation of systemic IL‐17 production. Circulating anti‐donor antibody (Ab) was not detected until CTLA4 Ig withdrawal. One monkey treated with donor Ag‐pulsed DCreg rejected its graft in association with progressively elevated anti‐donor Ab, 525 days posttransplant (160 days after withdrawal of immunosuppression). These findings indicate a modest but not statistically significant beneficial effect of donor Ag‐pulsed autologous DCreg infusion on NHP graft survival when administered with a minimal immunosuppressive drug regimen. 相似文献
17.
Novel Application of Localized Nanodelivery of Anti–Interleukin‐6 Protects Organ Transplant From Ischemia–Reperfusion Injuries 下载免费PDF全文
Z. Solhjou M. Uehara B. Bahmani O. H. Maarouf T. Ichimura C. R. Brooks W. Xu M. Yilmaz A. Elkhal S. G. Tullius I. Guleria M. M. McGrath R. Abdi 《American journal of transplantation》2017,17(9):2326-2337
Ischemia–reperfusion injury (IRI) evokes intragraft inflammatory responses, which markedly augment alloimmune responses against the graft. Understanding the mechanisms underlying these responses is fundamental to develop therapeutic regimens to prevent/ameliorate organ IRI. Here, we demonstrate that IRI results in a marked increase in mitochondrial damage and autophagy in dendritic cells (DCs). While autophagy is a survival mechanism for ischemic DCs, it also augments their production of interleukin (IL)‐6. Allograft‐derived dendritic cells (ADDCs) lacking autophagy‐related gene 5 (Atg5) showed higher death rates posttransplantation. Transplanted ischemic hearts from CD11cCre/Atg5 conditional knockout mice showed marked reduction in intragraft expression of IL‐6 compared with controls. To antagonize the effect of IL‐6 locally in the heart, we synthesized novel anti–IL‐6 nanoparticles with capacity for controlled release of anti–IL‐6 over time. Compared with systemic delivery of anti–IL‐6, localized delivery of anti–IL‐6 significantly reduced chronic rejection with a markedly lower amount administered. Despite improved allograft histology, there were no changes to splenic T cell populations, illustrating the importance of local IL‐6 in driving chronic rejection after IRI. These data carry potential clinical significance by identifying an innovative, targeted strategy to manipulate organs before transplantation to diminish inflammation, leading to improved long‐term outcomes. 相似文献
18.
Impact of Human Mutant TGFβ1/Fc Protein on Memory and Regulatory T Cell Homeostasis Following Lymphodepletion in Nonhuman Primates 下载免费PDF全文
H. Guo L. Lu R. Wang A. Perez‐Gutierrez H. S. Abdulkerim A. F. Zahorchak T. L. Sumpter K. A. Reimann A. W. Thomson M. B. Ezzelarab 《American journal of transplantation》2016,16(10):2994-3006
Transforming growth factor β1 (TGFβ1) plays a key role in T cell homeostasis and peripheral tolerance. We evaluated the influence of a novel human mutant TGFβ1/Fc (human IgG4 Fc) fusion protein on memory CD4+ and CD8+ T cell (Tmem) responses in vitro and their recovery following antithymocyte globulin (ATG)–mediated lymphodepletion in monkeys. TGFβ1/Fc induced Smad2/3 protein phosphorylation in rhesus and human peripheral blood mononuclear cells and augmented the suppressive effect of rapamycin on rhesus Tmem proliferation after either alloactivation or anti‐CD3/CD28 stimulation. In combination with IL‐2, the incidence of CD4+CD25hiFoxp3hi regulatory T cells (Treg) and Treg:Th17 ratios were increased. In lymphodepleted monkeys, whole blood trough levels of infused TGFβ1/Fc were maintained between 2 and 7 μg/mL for 35 days. Following ATG administration, total T cell numbers were reduced markedly. In those given TGFβ1/Fc infusion, CD8+ T cell recovery to predepletion levels was delayed compared to controls. Additionally, numbers of CD4+CD25hiCD127lo Treg increased at 4–6 weeks after depletion but subsequently declined to predepletion levels by 12 weeks. In all monkeys, CD4+CD25hiFoxp3hi Treg/CD4+IL‐17+ cell ratios were reduced, particularly after stopping TGFβ1/Fc infusion. Thus, human TGFβ1/Fc infusion may delay Tmem recovery following lymphodepletion in nonhuman primates. Combined (low‐dose) IL‐2 infusion may be required to improve the Treg:Th17 ratio following lymphodepletion. 相似文献
19.
B. Mahr N. Pilat S. Maschke N. Granofszky C. Schwarz L. Unger K. Hock A. M. Farkas C. Klaus H. Regele T. Wekerle 《American journal of transplantation》2017,17(12):3049-3059
Therapeutic administration of regulatory T cells (Tregs) leads to engraftment of conventional doses of allogeneic bone marrow (BM) in nonirradiated recipient mice conditioned with costimulation blockade and mammalian target of rapamycin inhibition. The mode of action responsible for this Treg effect is poorly understood but may encompass the control of costimulation blockade–resistant natural killer (NK) cells. We show that transient NK cell depletion at the time of BM transplantation led to BM engraftment and persistent chimerism without Treg transfer but failed to induce skin graft tolerance. In contrast, the permanent absence of anti–donor NK reactivity in mice grafted with F1 BM was associated with both chimerism and tolerance comparable to Treg therapy, implying that NK cell tolerization is a critical mechanism of Treg therapy. Indeed, NK cells of Treg‐treated BM recipients reshaped their receptor repertoire in the presence of donor MHC in a manner suggesting attenuated donor reactivity. These results indicate that adoptively transferred Tregs prevent BM rejection, at least in part, by suppressing NK cells and promote tolerance by regulating the appearance of NK cells expressing activating receptors to donor class I MHC. 相似文献
20.
Antoine Sicard Caroline Lamarche Madeleine Speck May Wong Isaac Rosado‐Snchez Mathilde Blois Nicolas Glaichenhaus Majid Mojibian Megan K. Levings 《American journal of transplantation》2020,20(6):1562-1573
Cell therapy with autologous donor‐specific regulatory T cells (Tregs) is a promising strategy to minimize immunosuppression in transplant recipients. Chimeric antigen receptor (CAR) technology has recently been used successfully to generate donor‐specific Tregs and overcome the limitations of enrichment protocols based on repetitive stimulations with alloantigens. However, the ability of CAR‐Treg therapy to control alloreactivity in immunocompetent recipients is unknown. We first analyzed the effect of donor‐specific CAR Tregs on alloreactivity in naive, immunocompetent mice receiving skin allografts. Tregs expressing an irrelevant or anti‐HLA‐A2‐specific CAR were administered to Bl/6 mice at the time of transplanting an HLA‐A2+ Bl/6 skin graft. Donor‐specific CAR‐Tregs, but not irrelevant‐CAR Tregs, significantly delayed skin rejection and diminished donor‐specific antibodies (DSAs) and frequencies of DSA‐secreting B cells. Donor‐specific CAR‐Treg–treated mice also had a weaker recall DSA response, but normal responses to an irrelevant antigen, demonstrating antigen‐specific suppression. When donor‐specific CAR Tregs were tested in HLA‐A2‐sensitized mice, they were unable to delay allograft rejection or diminish DSAs. The finding that donor‐specific CAR‐Tregs restrain de novo but not memory alloreactivity has important implications for their use as an adoptive cell therapy in transplantation. 相似文献