Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Am J Transplant ; 18(5): 1262-1269, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29377632

RESUMO

We have previously shown that 12 days of high-dose calcineurin inhibition induced tolerance in MHC inbred miniature swine receiving MHC-mismatched lung, kidney, or co-transplanted heart/kidney allografts. However, if lung grafts were procured from donation after brain death (DBD), and transplanted alone, they were rejected within 19-45 days. Here, we investigated whether donor brain death with or without allograft ischemia would also prevent tolerance induction in kidney or heart/kidney recipients. Four kidney recipients treated with 12 days of calcineurin inhibition received organs from donors rendered brain dead for 4 hours. Six heart/kidney recipients also treated with calcineurin inhibition received organs from donors rendered brain dead for 4 hours, 8 hours, or 4 hours with 4 additional hours of cold storage. In contrast to lung allograft recipients, all isolated kidney or heart/kidney recipients that received organs from DBD donors achieved long-term survival (>100 days) without histologic evidence of rejection. Proinflammatory cytokine gene expression was upregulated in lungs and hearts, but not kidney allografts, after brain death. These data suggest that the deleterious effects of brain death and ischemia on tolerance induction are organ-specific, which has implications for the application of tolerance to clinical transplantation.


Assuntos
Morte Encefálica/fisiopatologia , Rejeição de Enxerto/imunologia , Transplante de Coração , Isquemia/fisiopatologia , Transplante de Rim , Transplante de Pulmão , Tolerância ao Transplante/imunologia , Animais , Citocinas/genética , Citocinas/metabolismo , Sobrevivência de Enxerto , Especificidade de Órgãos , Suínos , Porco Miniatura , Doadores de Tecidos
2.
Am J Transplant ; 16(3): 979-86, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26469344

RESUMO

A 12-day course of high-dose tacrolimus induces tolerance of major histocompatibility complex-mismatched lung allografts in miniature swine but does not induce tolerance of heart allografts unless a kidney is cotransplanted. To determine whether lungs share with kidneys the ability to induce cardiac allograft tolerance, we investigated heart-lung cotransplantation using the same induction protocol. Hearts (n = 3), heart-kidneys (n = 3), lungs (n = 6), and hearts-lungs (n = 3) were transplanted into fully major histocompatibility complex-mismatched recipients treated with high-dose tacrolimus for 12 days. Serial biopsy samples were used to evaluate rejection, and in vitro assays were used to detect donor responsiveness. All heart-kidney recipients and five of six lung recipients demonstrated long-term graft survival for longer than 272 days, while all heart recipients rejected their allografts within 35 days. Tolerant recipients remained free of alloantibody and showed persistent donor-specific unresponsiveness by cell-mediated lympholysis/mixed-lymphocyte reaction. In contrast, heart-lung recipients demonstrated rejection of both allografts (days 47, 55, and 202) and antidonor responsiveness in vitro. In contrast to kidneys, lung cotransplantation leads to rejection of both heart and lung allografts, indicating that lungs do not have the same tolerogenic capacity as kidneys. We conclude that cells or cell products present in kidney, but not heart or lung allografts, have a unique capacity to confer unresponsiveness on cotransplanted organs, most likely by amplifying host regulatory mechanisms.


Assuntos
Rejeição de Enxerto/imunologia , Transplante de Coração , Tolerância Imunológica/imunologia , Transplante de Pulmão , Complexo Principal de Histocompatibilidade/imunologia , Complicações Pós-Operatórias , Tolerância ao Transplante/imunologia , Animais , Sobrevivência de Enxerto , Imunossupressores/uso terapêutico , Teste de Cultura Mista de Linfócitos , Suínos , Porco Miniatura
3.
Am J Transplant ; 15(12): 3055-66, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26190648

RESUMO

Tolerance of allografts achieved in mice via stable mixed hematopoietic chimerism relies essentially on continuous elimination of developing alloreactive T cells in the thymus (central deletion). Conversely, while only transient mixed chimerism is observed in nonhuman primates and patients, it is sufficient to ensure tolerance of kidney allografts. In this setting, it is likely that tolerance depends on peripheral regulatory mechanisms rather than thymic deletion. This implies that, in primates, upsetting the balance between inflammatory and regulatory alloimmunity could abolish tolerance and trigger the rejection of previously accepted renal allografts. In this study, six monkeys that were treated with a mixed chimerism protocol and had accepted a kidney allograft for periods of 1-10 years after withdrawal of immunosuppression received subcutaneous injections of IL-2 cytokine (0.6-3 × 10(6) IU/m(2) ). This resulted in rapid rejection of previously tolerated renal transplants and was associated with an expansion and reactivation of alloreactive pro-inflammatory memory T cells in the host's lymphoid organs and in the graft. This phenomenon was prevented by anti-CD8 antibody treatment. Finally, this process was reversible in that cessation of IL-2 administration aborted the rejection process and restored normal kidney graft function.


Assuntos
Rejeição de Enxerto/etiologia , Interleucina-2/administração & dosagem , Falência Renal Crônica/cirurgia , Transplante de Rim , Complicações Pós-Operatórias , Quimeras de Transplante/imunologia , Tolerância ao Transplante/imunologia , Animais , Transplante de Medula Óssea , Quimerismo , Taxa de Filtração Glomerular , Rejeição de Enxerto/tratamento farmacológico , Sobrevivência de Enxerto , Injeções Subcutâneas , Testes de Função Renal , Macaca fascicularis , Camundongos , Fatores de Risco , Condicionamento Pré-Transplante , Transplante Homólogo
4.
Am J Transplant ; 15(8): 2231-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25904524

RESUMO

While the induction of transient mixed chimerism has tolerized MHC-mismatched renal grafts in nonhuman primates and patients, this approach has not been successful for more immunogenic organs. Here, we describe a modified delayed-tolerance-induction protocol resulting in three out of four monkeys achieving long-term lung allograft survival without ongoing immunosuppression. Two of the tolerant monkeys displayed stable mixed lymphoid chimerism, and the other showed transient chimerism. Serial biopsies and post-mortem specimens from the tolerant monkeys revealed no signs of chronic rejection. The tolerant recipients also exhibited T cell unresponsiveness and a lack of alloantibody. This is the first report of durable mixed chimerism and successful tolerance induction of MHC-mismatched lungs in primates.


Assuntos
Quimerismo , Hematopoese , Transplante de Pulmão , Animais , Macaca fascicularis , Transplante Homólogo
5.
Am J Transplant ; 15(5): 1415-20, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25772308

RESUMO

Despite advances in surgical technique and clinical care, lung transplantation still remains a short-term solution for the treatment of end-stage lung disease. To date, there has been limited experience in experimental lung transplantation using nonhuman primate models. Therefore, we have endeavored to develop a long-term, nonhuman primate model of orthotopic lung transplantation for the ultimate purpose of designing protocols to induce tolerance of lung grafts. Here, we report our initial results in developing this model and our observation that the nonhuman primate lung is particularly prone to rejection. This propensity toward rejection may be a consequence of 1) upregulated nonspecific inflammation, and 2) a larger number of pre-existing alloreactive memory T cells, leading to augmented deleterious immune responses. Our data show that triple-drug immunosuppression mimicking clinical practice is not sufficient to prevent acute rejection in nonhuman primate lung transplantation. The addition of horse-derived anti-thymocyte globulin and a monoclonal antibody to the IL-6 receptor allowed six out of six lung recipients to be free of rejection for over 120 days.


Assuntos
Pneumopatias/cirurgia , Transplante de Pulmão , Animais , Soro Antilinfocitário/química , Teste de Histocompatibilidade , Cavalos , Tolerância Imunológica , Memória Imunológica/imunologia , Terapia de Imunossupressão , Inflamação/imunologia , Pulmão/patologia , Macaca fascicularis , Complexo Principal de Histocompatibilidade , Modelos Animais , Receptores de Interleucina-6/metabolismo , Linfócitos T/citologia , Transplante Autólogo , Transplante Homólogo
6.
Am J Transplant ; 15(6): 1580-90, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25824550

RESUMO

Kidney allografts possess the ability to enable a short course of immunosuppression to induce tolerance of themselves and of cardiac allografts across a full-MHC barrier in miniature swine. However, the renal element(s) responsible for kidney-induced cardiac allograft tolerance (KICAT) are unknown. Here we investigated whether MHC disparities between parenchyma versus hematopoietic-derived "passenger" cells of the heart and kidney allografts affected KICAT. Heart and kidney allografts were co-transplanted into MHC-mismatched recipients treated with high-dose tacrolimus for 12 days. Group 1 animals (n = 3) received kidney and heart allografts fully MHC-mismatched to each other and to the recipient. Group 2 animals (n = 3) received kidney and heart allografts MHC-matched to each other but MHC-mismatched to the recipient. Group 3 animals (n = 3) received chimeric kidney allografts whose parenchyma was MHC-mismatched to the donor heart. Group 4 animals (n = 3) received chimeric kidney allografts whose passenger leukocytes were MHC-mismatched to the donor heart. Five of six heart allografts in Groups 1 and 3 rejected <40 days. In contrast, heart allografts in Groups 2 and 4 survived >150 days without rejection (p < 0.05). These data demonstrate that KICAT requires MHC-matching between kidney allograft parenchyma and heart allografts, suggesting that cells intrinsic to the kidney enable cardiac allograft tolerance.


Assuntos
Transplante de Coração , Coração/fisiologia , Histocompatibilidade/fisiologia , Transplante de Rim , Rim/fisiologia , Complexo Principal de Histocompatibilidade/fisiologia , Tolerância ao Transplante/fisiologia , Aloenxertos , Animais , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Histocompatibilidade/imunologia , Terapia de Imunossupressão , Imunossupressores/uso terapêutico , Complexo Principal de Histocompatibilidade/imunologia , Modelos Animais , Suínos , Porco Miniatura , Tacrolimo/uso terapêutico , Obtenção de Tecidos e Órgãos , Tolerância ao Transplante/imunologia
7.
Am J Transplant ; 14(12): 2704-12, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25394378

RESUMO

We have previously reported successful induction of renal allograft tolerance via a mixed chimerism approach in nonhuman primates. In those studies, we found that costimulatory blockade with anti-CD154 mAb was an effective adjunctive therapy for induction of renal allograft tolerance. However, since anti-CD154 mAb is not clinically available, we have evaluated CTLA4Ig as an alternative agent for effecting costimulation blockade in this treatment protocol. Two CTLA4Igs, abatacept and belatacept, were substituted for anti-CD154 mAb in the conditioning regimen (low dose total body irradiation, thymic irradiation, anti-thymocyte globulin and a 1-month posttransplant course of cyclosporine [CyA]). Three recipients treated with the abatacept regimen failed to develop comparable lymphoid chimerism to that achieved with anti-CD154 mAb treatment and these recipients rejected their kidney allografts early. With the belatacept regimen, four of five recipients developed chimerism and three of these achieved long-term renal allograft survival (>861, >796 and >378 days) without maintenance immunosuppression. Neither chimerism nor long-term allograft survival were achieved in two recipients treated with the belatacept regimen but with a lower, subtherapeutic dose of CyA. This study indicates that CD28/B7 blockade with belatacept can provide a clinically applicable alternative to anti-CD154 mAb for promoting chimerism and renal allograft tolerance.


Assuntos
Transplante de Medula Óssea , Quimerismo , Imunoconjugados/administração & dosagem , Imunossupressores/administração & dosagem , Nefropatias/imunologia , Transplante de Rim , Tolerância ao Transplante/imunologia , Abatacepte , Animais , Anticorpos Monoclonais/administração & dosagem , Citometria de Fluxo , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/imunologia , Nefropatias/terapia , Testes de Função Renal , Macaca fascicularis , Doadores de Tecidos , Quimeras de Transplante/imunologia , Condicionamento Pré-Transplante , Transplante Homólogo , Irradiação Corporal Total
8.
Am J Transplant ; 14(10): 2263-2274, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25155089

RESUMO

The relative contribution of central and peripheral mechanisms to the generation and maintenance of allograft tolerance is of considerable interest. Here, we present new evidence that regulatory T cells (Foxp3(+) ) maintain skin and heart allograft tolerance in mixed hematopoietic chimeric mice. Transient depletion of both donor- and recipient-derived Foxp3(+) cells was necessary and sufficient to induce decisive rejection of long-accepted skin and heart allografts. In contrast, stable hematopoietic chimerism remained, and there was no detectable induction of donor-specific reactivity to hematopoietic cells. Foxp3(+) cell depletion did not result in the rejection of skin grafts of only MHC-disparate donors (B6.C-H2(d) /bByJ), indicating that MHC antigens were not the target in the graft. We conclude that two different mechanisms of tolerance are present in mixed chimeras. Hematopoietic chimerism, resistant to Foxp3(+) depletion, is probably due to deletional tolerance to MHC antigens, as supported by previous studies. In contrast, regulatory tolerance mechanisms involving Foxp3(+) cells are required to control reactivity against non-MHC antigens not present on hematopoietic lineages.


Assuntos
Quimera , Fatores de Transcrição Forkhead/imunologia , Transplante de Coração , Tolerância Imunológica , Depleção Linfocítica , Transplante de Pele , Linfócitos T/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Rejeição de Enxerto/imunologia , Camundongos
9.
Am J Transplant ; 13(10): 2558-66, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24033991

RESUMO

We have previously shown that tolerance of kidney allografts across a full major histocompatibility complex (MHC) barrier can be induced in miniature swine by a 12-day course of high-dose tacrolimus. However, that treatment did not prolong survival of heart allografts across the same barrier. We have now tested the effect of cotransplanting an allogeneic heart and kidney from the same MHC-mismatched donor using the same treatment regimen. Heart allografts (n = 3) or heart plus kidney allografts (n = 5) were transplanted into MHC-mismatched recipients treated with high-dose tacrolimus for 12 days. As expected, all isolated heart allografts rejected by postoperative day 40. In contrast, heart and kidney allografts survived for >200 days with no evidence of rejection on serial cardiac biopsies. Heart/kidney recipients lost donor-specific responsiveness in cell-mediated lympholysis and mixed-lymphocyte reaction assays, were free of alloantibody and exhibited prolonged survival of donor, but not third-party skin grafts. Late (>100 days) removal of the kidney allografts did not cause acute rejection of the heart allografts (n = 2) and did not abrogate donor-specific unresponsiveness in vitro. While kidney-induced cardiac allograft tolerance (KICAT) has previously been demonstrated across a Class I disparity, these data demonstrate that this phenomenon can also be observed across the more clinically relevant full MHC mismatch. Elucidating the renal element(s) responsible for KICAT could provide mechanistic information relevant to the induction of tolerance in recipients of isolated heart allografts as well as other tolerance-resistant organs.


Assuntos
Rejeição de Enxerto/imunologia , Transplante de Coração , Transplante de Rim , Complexo Principal de Histocompatibilidade/imunologia , Doadores de Tecidos , Tolerância ao Transplante , Aloenxertos , Animais , Citometria de Fluxo , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Imunossupressores , Transplante de Pele , Suínos , Porco Miniatura
10.
Am J Transplant ; 12(2): 313-21, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22070565

RESUMO

Chronic allograft vasculopathy (CAV) in murine heart allografts can be elicited by adoptive transfer of donor specific antibody (DSA) to class I MHC antigens and is independent of complement. Here we address the mechanism by which DSA causes CAV. B6.RAG1(-/-) or B6.RAG1(-/-)C3(-/-) (H-2(b)) mice received B10.BR (H-2(k)) heart allografts and repeated doses of IgG2a, IgG1 or F(ab')(2) fragments of IgG2a DSA (anti-H-2(k)). Intact DSA regularly elicited markedly stenotic CAV in recipients over 28 days. In contrast, depletion of NK cells with anti-NK1.1 reduced significantly DSA-induced CAV, as judged morphometrically. Recipients genetically deficient in mature NK cells (γ-chain knock out) also showed decreased severity of DSA-induced CAV. Direct NK reactivity to the graft was not necessary. F(ab')(2) DSA fragments, even at doses twofold higher than intact DSA, were inactive. Graft microvascular endothelial cells responded to DSA in vivo by increased expression of phospho-extracellular signal-regulated kinase (pERK), a response not elicited by F(ab')(2) DSA. We conclude that antibody mediates CAV through NK cells, by an Fc dependent manner. This new pathway adds to the possible mechanisms of chronic rejection and may relate to the recently described C4d-negative chronic antibody-mediated rejection in humans.


Assuntos
Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Imunidade Celular , Isoanticorpos/imunologia , Células Matadoras Naturais/imunologia , Animais , Doença Crônica , Modelos Animais de Doenças , Citometria de Fluxo , Rejeição de Enxerto/patologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante Homólogo
11.
Am J Transplant ; 12(5): 1290-5, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22300400

RESUMO

We have previously shown that a short course of high-dose tacrolimus induces long-term tolerance to fully mismatched lung allografts procured from healthy MHC-inbred miniature swine. Here, we investigate whether donor brain death affects tolerance induction. Four recipient swine were transplanted with fully mismatched lung grafts from donors that were rendered brain dead and mechanically ventilated for 4 h before procurement (Group 1). These recipients were compared to two control groups (Group 2: 4 h of donor ventilation without brain death [n = 5]; and Group 3: no donor brain death with <1 h of ventilation [n = 6]). All recipients were treated with a 12-day course of tacrolimus. In contrast to both groups of control animals, the swine transplanted with lung allografts from brain dead donors all rejected their grafts by postoperative day 45 and showed persistent responsiveness to donor antigen by MLR. Several additional swine underwent brain death induction and/or mechanical ventilation alone to determine the effects of these procedures on the expression of proinflammatory molecules. Significant increases in serum concentrations of IL-1, TNF-α and IL-10 were seen after brain death. Upregulation of IL-1 and IL-6 gene expression was also observed.


Assuntos
Morte Encefálica/imunologia , Rejeição de Enxerto/imunologia , Tolerância Imunológica/imunologia , Transplante de Pulmão/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Doadores de Tecidos , Animais , Ensaio de Imunoadsorção Enzimática , Interleucina-1/genética , Interleucina-1/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Modelos Animais , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Suínos , Porco Miniatura , Transplante Homólogo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
Am J Transplant ; 12(9): 2532-7, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22682297

RESUMO

IL-2 is a known potent T cell growth factor that amplifies lymphocyte responses in vivo. This capacity has led to the use of high-dose IL-2 to enhance T cell immunity in patients with AIDS or cancer. However, more recent studies have indicated that IL-2 is also critical for the development and peripheral expansion of regulatory T cells (Tregs). In the current study, low-dose IL-2 (1 million IU/m(2) BSA/day) was administered to expand Tregs in vivo in naïve nonhuman primates. Our study demonstrated that low-dose IL-2 therapy significantly expanded peripheral blood CD4(+) and CD8(+) Tregs in vivo with limited expansion of non-Treg cells. These expanded Tregs are mainly CD45RA(-) Foxp3(high) activated Tregs and demonstrated potent immunosuppressive function in vitro. The results of this preclinical study can serve as a basis to develop Treg immunotherapy, which has significant therapeutic potential in organ/cellular transplantation.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Interleucina-2/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Relação Dose-Resposta a Droga , Interleucina-2/administração & dosagem , Macaca fascicularis , Masculino , Linfócitos T Reguladores/imunologia
13.
Am J Transplant ; 12(2): 330-40, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22053723

RESUMO

The presence of alloreactive memory T cells is a major barrier for induction of tolerance in primates. In theory, delaying conditioning for tolerance induction until after organ transplantation could further decrease the efficacy of the regimen, since preexisting alloreactive memory T cells might be stimulated by the transplanted organ. Here, we show that such "delayed tolerance" can be induced in nonhuman primates through the mixed chimerism approach, if specific modifications to overcome/avoid donor-specific memory T-cell responses are provided. These modifications include adequate depletion of CD8+ memory T cells and timing of donor bone marrow administration to minimize levels of proinflammatory cytokines. Using this modified approach, mixed chimerism was induced successfully in 11 of 13 recipients of previously placed renal allografts and long-term survival without immunosuppression could be achieved in at least 6 of these 11 animals.


Assuntos
Transplante de Medula Óssea/imunologia , Sobrevivência de Enxerto/imunologia , Memória Imunológica/imunologia , Transplante de Rim/imunologia , Linfócitos T/imunologia , Quimeras de Transplante/imunologia , Tolerância ao Transplante/imunologia , Animais , Transplante de Medula Óssea/patologia , Modelos Animais de Doenças , Citometria de Fluxo , Seguimentos , Transplante de Rim/patologia , Macaca fascicularis , Masculino , Condicionamento Pré-Transplante/métodos , Transplante Homólogo/imunologia , Transplante Homólogo/patologia
14.
Am J Transplant ; 10(3): 510-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20055805

RESUMO

Complement fixation, as evidenced by C4d in the microvasculature, is a widely accepted criterion of antibody-mediated rejection. Complement fixation has been shown to be essential in acute antibody-mediated rejection, but its role in chronic rejection has not been addressed. Previous studies showed that passive transfer of complement fixing monoclonal IgG2a anti-H-2Kk into B6.RAG1-/- KO recipients of B10.BR hearts led to progressive chronic transplant arteriopathy (CTA) over 14-28 days, accompanied by C4d deposition. The present studies were designed to test whether complement was required for these lesions. We report that a noncomplement fixing donor-specific alloantibody (DSA, monoclonal IgG1 anti-H-2Kk) injected into B6.RAG1-/- KO recipients of B10.BR hearts also promotes CTA, without C4d deposition. Furthermore, a passive transfer of DSA (monoclonal IgG2a anti-H-2Kk) initiated endarteritis followed by CTA in B6.RAG1-/- mice genetically deficient in the third component of complement (RAG1-/-C3-/-). These studies indicate that antibody to class I MHC antigens can trigger chronic arterial lesions in vivo without complement participation, in contrast to acute antibody-mediated rejection. This pathway may be relevant to C4d-negative chronic rejection sometimes observed in patients with DSA, and argues that lack of C4d deposition does not exclude antibody-mediated chronic rejection.


Assuntos
Anticorpos/química , Artérias/patologia , Complemento C4b/genética , Proteínas do Sistema Complemento/metabolismo , Endarterite/imunologia , Transplante de Coração/métodos , Fragmentos de Peptídeos/genética , Animais , Anticorpos Monoclonais/metabolismo , Progressão da Doença , Coração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Tempo
15.
Am J Transplant ; 10(6): 1375-84, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20486921

RESUMO

The high frequency of memory T cells present in primates is thought to represent a major barrier to tolerance induction in transplantation. Therefore, it is crucial to characterize these memory T cells and determine their functional properties. High numbers of memory T cells were detected in peripheral blood and all lymphoid tissues except lymph nodes, which were essentially the site of naïve T cells. The majority of CD8(+) memory T cells were effector memory cells located in the blood and bone marrow while most CD4(+) memory T cells were central memory cells present in the spleen. Next, memory T cells from over 100 monkeys were tested for their response to alloantigens by ELISPOT. Memory alloreactivity mediated via direct but not indirect allorecognition was detected in all animals. The frequency of allospecific memory T cells varied dramatically depending upon the nature of the responder/stimulator monkey combination tested. MHC gene matching was generally associated with a low-memory alloreactivity. Nevertheless, low anamnestic alloresponses were also found in a significant number of fully MHC-mismatched monkey combinations. These results show that selected donor/recipient combinations displaying a low memory alloresponsiveness can be found. These combinations may be more favorable for transplant tolerance induction.


Assuntos
Linfócitos T/imunologia , Animais , Memória Imunológica/imunologia , Isoantígenos/imunologia , Macaca fascicularis , Masculino
16.
Am J Transplant ; 9(11): 2479-84, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19843029

RESUMO

Viral infections including those due to cytomegalovirus have been associated with accelerated cardiac allograft vasculopathy (CAV) in clinical trials and some animal models. Evidence demonstrating a direct causal relationship between such infections and de novo formation of coronary vascular lesions is lacking. Heterotopic murine cardiac transplants were performed in a parental to F1 combination in animals lacking both T- and B-lymphocytes (RAG(-/-)). Coronary vasculopathy developed almost exclusively in the presence of recipient infection with lymphocytic choriomeningitis virus but not in uninfected controls. This process was also dependent upon the presence of natural killer (NK) cells as depletion of NK cells abrogated the process. These data show that a viral infection in its native host, and not previously implicated in the production of CAV, can contribute to the development of advanced coronary vascular lesions in cardiac allotransplants in mice. These data also suggest that virus-induced CAV can develop via an NK-cell-dependent pathway in the absence of T- and B-lymphocytes.


Assuntos
Infecções por Arenaviridae/imunologia , Doença das Coronárias/virologia , Transplante de Coração , Hospedeiro Imunocomprometido , Células Matadoras Naturais/virologia , Vírus da Coriomeningite Linfocítica , Animais , Infecções por Arenaviridae/complicações , Linfócitos B/imunologia , Doença das Coronárias/imunologia , Sobrevivência de Enxerto/imunologia , Proteínas de Homeodomínio/genética , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Linfócitos T/imunologia , Imunologia de Transplantes , Transplante Homólogo
17.
Am J Transplant ; 9(1): 105-13, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19145702

RESUMO

We studied the effects of indirect allorecognition on the induction and maintenance phases of tolerance in miniature swine cotransplanted with heart and kidney allografts. MHC class I-mismatched heart and kidney grafts were cotransplanted in recipients receiving CyA for 12 days. Recipients were unimmunized or immunized with a set of donor-derived or control third-party MHC class I peptides either 21 days prior to transplantation or over 100 days after transplantation. T-cell proliferation, delayed type hypersensitivity reaction (DTH) and antibody production were assessed. All animals injected with donor MHC class I peptides developed potent indirect alloresponses specific to the immunizing peptides. While untreated recipients developed stable tolerance, all animals preimmunized with donor allopeptides rejected kidney-heart transplants acutely. In contrast, when peptide immunization was delayed until over 100 days after kidney-heart transplantation, no effects were observed on graft function or in vitro measures of alloimmunity. Donor peptide immunization prevented tolerance when administered to recipients pre transplantation but did not abrogate tolerance when administered to long-term survivors post transplantation. This suggests that the presence of T cells activated via indirect allorecognition represent a barrier to the induction but not the maintenance of tolerance.


Assuntos
Transplante de Coração/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Tolerância Imunológica , Transplante de Rim/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Hipersensibilidade Tardia , Suínos , Porco Miniatura , Transplante Homólogo
18.
Science ; 202(4364): 217-9, 1978 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-694527

RESUMO

Response saturation of blue-sensitive cone pathways was studied by measuring increment thresholds for violet test flashes on flashed violet fields in the presence of a steady yellow "auxiliary" field of constant radiance. Adding intense yellow field flashes to the violet field flash could eliminate or reduce response saturation (greatly reduce threshold), whereas "negative" yellow field flashes drove the mechanism to further saturation. The response saturation is thus not, in general, controlled exclusively by independent blue-sensitive cones but by spectrally opponent mechanisms that receive opposite-signed signals from blue-sensitive cones and from green-or red-sensitive cones. These results add to a growing number of studies that demonstrate that detection of signals from blue-sensitive cones is largely through a color-opponent pathway.


Assuntos
Percepção de Cores/fisiologia , Células Fotorreceptoras/fisiologia , Visão Ocular , Potenciais de Ação , Humanos
19.
Science ; 207(4430): 555-7, 1980 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-7352270

RESUMO

Color-selective spatial adaptation of the short-wavelength, or blue-sensitive, pathway was demonstrated. The adaptation was orientation selective and strongly monocular. Adaptation was assessed by measuring visibility thresholds for monochromatic gratings in subjects adapted to high-contrast violet gratings designed to stimulate only blue-sensitive cones. The results showed spatially selective, adaptable channels within the short-wavelength pathway.


Assuntos
Adaptação Fisiológica , Percepção de Cores/fisiologia , Células Fotorreceptoras/fisiologia , Percepção de Forma/fisiologia , Humanos , Percepção Visual/fisiologia
20.
Transplant Proc ; 41(1): 429-30, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19249572

RESUMO

BACKGROUND: We have previously reported the successful induction of renal allograft tolerance in non-human primates using a nonmyeloablative conditioning regimen to produce a mixed-chimeric state in the recipient. In the present study, we applied this same technique to lung allotransplantation in cynomolgus monkeys. METHODS: Nine pairs of fully major histocompatibility complex (MHC)-mismatched cynomolgus monkeys were used. The conditioning regimen consisted of total body irradiation, thymic irradiation, and antithymocyte globulin. The recipients underwent lung and bone marrow transplantation, followed by anti-CD154 monoclonal antibody (mAb), and a 1-month course of cyclosporine. The regimen included anti-CD8 mAb in the last 5 recipients and alpha 1-antitripsin in the last 3 recipients. The results were compared with 8 recipients that received kidney allografts using the same regimen. RESULTS: Transient chimerism developed in all lung recipients, as was previously seen in the kidney recipients. Nonetheless, the lung recipients rejected their allografts significant earlier than the kidney recipients (P < .01). CONCLUSIONS: Despite the successful induction of mixed chimerism in recipients of fully MHC-mismatched lung allografts, we have not observed long-term graft survival, as has been seen in an analogous kidney model. Strategies to overcome this problem include organ-specific modifications of the transplant regimen.


Assuntos
Sobrevivência de Enxerto/imunologia , Tolerância Imunológica , Transplante de Rim/imunologia , Transplante de Pulmão/imunologia , Quimeras de Transplante , Sistema ABO de Grupos Sanguíneos , Animais , Soro Antilinfocitário/uso terapêutico , Transplante de Medula Óssea/imunologia , Teste de Histocompatibilidade , Macaca fascicularis , Complexo Principal de Histocompatibilidade/imunologia , Masculino , Irradiação Corporal Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA