Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 71(10): 2421-2431, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35237846

RESUMEN

Ipilimumab, a monoclonal antibody that recognizes cytotoxic T-lymphocyte associated protein 4 (CTLA-4), was the first immune checkpoint inhibitor approved by the FDA to treat metastatic melanoma patients. Multiple preclinical studies have proposed that Fc effector functions of anti-CTLA-4 therapy are required for anti-tumor efficacy, in part, through the depletion of intratumoral regulatory T cells (Tregs). However, the contribution of the Fc-independent functions of anti-CTLA-4 antibodies to the observed efficacy is not fully understood. H11, a non-Fc-containing single-domain antibody (VHH) against CTLA-4, has previously been demonstrated to block CTLA-4-ligand interaction. However, in vivo studies demonstrated lack of anti-tumor efficacy with H11 treatment. Here, we show that a half-life extended H11 (H11-HLE), despite the lack of Fc effector functions, induced potent anti-tumor efficacy in mouse syngeneic tumor models. In addition, a non-Fc receptor binding version of ipilimumab (Ipi-LALAPG) also demonstrated anti-tumor activity in the absence of Treg depletion. Thus, we demonstrate that Fc-independent functions of anti-CTLA-4 antibodies contributed to anti-tumor efficacy, which may indicate that non-Treg depleting activity of anti-CTLA-4 therapy could benefit cancer patients in the clinic.


Asunto(s)
Melanoma , Linfocitos T Reguladores , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígeno CTLA-4 , Modelos Animales de Enfermedad , Ipilimumab/farmacología , Ipilimumab/uso terapéutico , Melanoma/tratamiento farmacológico , Ratones
2.
Front Immunol ; 8: 80, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28210263

RESUMEN

B lymphocytes contribute to acute and chronic allograft rejection through their production of donor-specific antibodies (DSAs). In addition, B cells present allopeptides bound to self-MHC class II molecules and provide costimulation signals to T cells, which are essential to their activation and differentiation into memory T cells. On the other hand, both in laboratory rodents and patients, the concept of effector T cell regulation by B cells is gaining traction in the field of transplantation. Specifically, clinical trials using anti-CD20 monoclonal antibodies to deplete B cells and reverse DSA had a deleterious effect on rates of acute cellular rejection; a peculiar finding that calls into question a central paradigm in transplantation. Additional work in humans has characterized IL-10-producing B cells (IgM memory and transitional B cells), which suppress the proliferation and inflammatory cytokine productions of effector T cells in vitro. Understanding the mechanisms of regulating the alloresponse is critical if we are to achieve operational tolerance across transplantation. This review will focus on recent evidence in murine and human transplantation with respect to non-traditional roles for B cells in determining clinical outcomes.

3.
Sci Immunol ; 1(1)2016 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-27942611

RESUMEN

Transplantation of allogeneic organs and tissues represents a lifesaving procedure for a variety of patients affected with end-stage diseases. Although current immunosuppressive therapy prevents early acute rejection, it is associated with nephrotoxicity and increased risks for infection and neoplasia. This stresses the need for selective immune-based therapies relying on manipulation of lymphocyte recognition of donor antigens. The passenger leukocyte theory states that allograft rejection is initiated by recipient T cells recognizing donor major histocompatibility complex (MHC) molecules displayed on graft leukocytes migrating to the host's lymphoid organs. We revisited this concept in mice transplanted with allogeneic skin, heart, or islet grafts using imaging flow cytometry. We observed no donor cells in the lymph nodes and spleen of skin-grafted mice, but we found high numbers of recipient cells displaying allogeneic MHC molecules (cross-dressed) acquired from donor microvesicles (exosomes). After heart or islet transplantation, we observed few donor leukocytes (100 per million) but large numbers of recipient cells cross-dressed with donor MHC (>90,000 per million). Last, we showed that purified allogeneic exosomes induced proinflammatory alloimmune responses by T cells in vitro and in vivo. Collectively, these results suggest that recipient antigen-presenting cells cross-dressed with donor MHC rather than passenger leukocytes trigger T cell responses after allotransplantation.

4.
Neurol Neuroimmunol Neuroinflamm ; 3(5): e276, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27648463

RESUMEN

Neuromyelitis optica (NMO) and spectrum disorder (NMO/SD) represent a vexing process and its clinical variants appear to have at their pathogenic core the loss of immune tolerance to the aquaporin-4 water channel protein. This process results in a characteristic pattern of astrocyte dysfunction, loss, and demyelination that predominantly affects the spinal cord and optic nerves. Although several empirical therapies are currently used in the treatment of NMO/SD, none has been proven effective in prospective, adequately powered, randomized trials. Furthermore, most of the current therapies subject patients to long-term immunologic suppression that can cause serious infections and development of cancers. The following is the first of a 2-part description of several key immune mechanisms in NMO/SD that might be amenable to therapeutic restoration of immune tolerance. It is intended to provide a roadmap for how potential immune tolerance restorative techniques might be applied to patients with NMO/SD. This initial installment provides a background rationale underlying attempts at immune tolerization. It provides specific examples of innovative approaches that have emerged recently as a consequence of technical advances. In several autoimmune diseases, these strategies have been reduced to practice. Therefore, in theory, the identification of aquaporin-4 as the dominant autoantigen makes NMO/SD an ideal candidate for the development of tolerizing therapies or cures for this increasingly recognized disease.

5.
Neurol Neuroimmunol Neuroinflamm ; 3(5): e277, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27648464

RESUMEN

Neuromyelitis optica spectrum disorder (NMO/SD) and its clinical variants have at their core the loss of immune tolerance to aquaporin-4 and perhaps other autoantigens. The characteristic phenotype is disruption of astrocyte function and demyelination of spinal cord, optic nerves, and particular brain regions. In this second of a 2-part article, we present further perspectives regarding the pathogenesis of NMO/SD and how this disease might be amenable to emerging technologies aimed at restoring immune tolerance to disease-implicated self-antigens. NMO/SD appears to be particularly well-suited for these strategies since aquaporin-4 has already been identified as the dominant autoantigen. The recent technical advances in reintroducing immune tolerance in experimental models of disease as well as in humans should encourage quantum leaps in this area that may prove productive for novel therapy. In this part of the article series, the potential for regulatory T and B cells is brought into focus, as are new approaches to oral tolerization. Finally, a roadmap is provided to help identify potential issues in clinical development and guide applications in tolerization therapy to solving NMO/SD through the use of emerging technologies. Each of these perspectives is intended to shine new light on potential cures for NMO/SD and other autoimmune diseases, while sparing normal host defense mechanisms.

6.
J Clin Invest ; 125(10): 3751-3, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26389671

RESUMEN

Immune-suppressive cell populations, including Tregs and suppressor monocytes, have been implicated in long-term survival of allografts in both human transplant recipients and animal models. The factors that drive differentiation and function of these cell populations are not completely understood. In this issue, Bézie and colleagues identify IL-34 as an important mediator of allograft tolerance in a rat model of heart transplantation. Their data support a model in which IL-34 production by Tregs promotes a population of suppressive macrophages that in turn promote Treg differentiation. The results of this study support further exploration of the immunosuppressive properties of IL-34.


Asunto(s)
Interleucinas/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante , Animales , Humanos , Masculino
7.
Curr Opin Organ Transplant ; 20(4): 369-75, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26107970

RESUMEN

PURPOSE OF REVIEW: B cells are known to play a central role in humoral immunity and to boost cellular immunity, however, in a variety of experimental models, B-cell subsets ameliorate inflammation and autoimmune disease, indicating that they can also play a regulatory role. Here, we highlight the advances in regulatory B-cell (Breg) biology of the past year with an emphasis on findings pertinent to transplantation. Several recent observations highlight the relevance to clinical transplantation. Data from at least three independent groups demonstrated that spontaneously tolerant renal transplant recipients exhibit a peripheral blood B-cell signature although the significance of these data remains unclear. Moreover, new data suggest that regulatory B cells may serve as a biomarker for long-term allograft outcomes. Finally, recent evidence suggesting that plasma cells may be an essential component of Bregs raises new concerns about targeting antibody producing cells. RECENT FINDINGS: We describe new information on Breg mechanisms of action to suppress the alloresponse, signals to expand Bregs in vitro, and more functional evidence of Breg involvement in operationally tolerant kidney patients and in maintaining stable allograft function. SUMMARY: Although lymphocyte depletion remains central to tolerance induction therapy, the sparing or expansion of regulatory B cells may be an additional strategy to preempt graft rejection.


Asunto(s)
Linfocitos B/inmunología , Tolerancia Inmunológica , Animales , Rechazo de Injerto/inmunología , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral , Trasplante Homólogo
8.
Cell Transplant ; 24(8): 1599-1614, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24612609

RESUMEN

The role of the novel costimulatory molecule TIM4 in anti-islet response is unknown. We explored TIM4 expression and targeting in Th1 (BALB/c islets into C57BL/6 mice) and Th2 (BALB/c islets into Tbet(-/-) C57BL/6 mice) models of anti-islet alloimmune response and in a model of anti-islet autoimmune response (diabetes onset in NOD mice). The targeting of TIM4, using the monoclonal antibody RMT4-53, promotes islet graft survival in a Th1 model, with 30% of the graft surviving in the long term; islet graft protection appears to be mediated by a Th1 to Th2 skewing of the immune response. Differently, in the Th2 model, TIM4 targeting precipitates graft rejection by further enhancing the Th2 response. The effect of anti-TIM4 treatment in preventing autoimmune diabetes was marginal with only minor Th1 to Th2 skewing. B-Cell depletion abolished the effect of TIM4 targeting. TIM4 is expressed on human B-cells and is upregulated in diabetic and islet-transplanted patients. Our data suggest a model in which TIM4 targeting promotes Th2 response over Th1 via B-cells. The targeting of TIM4 could become a component of an immunoregulatory protocol in clinical islet transplantation, aiming at redirecting the immune system toward a Th2 response.


Asunto(s)
Autoinmunidad , Trasplante de Islotes Pancreáticos/inmunología , Proteínas de la Membrana/metabolismo , Células Th2/inmunología , Adulto , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Diferenciación Celular , Citocinas/análisis , Citocinas/metabolismo , Diabetes Mellitus Experimental/mortalidad , Diabetes Mellitus Experimental/cirugía , Diabetes Mellitus Tipo 1/cirugía , Femenino , Supervivencia de Injerto/inmunología , Humanos , Islotes Pancreáticos/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Persona de Mediana Edad , Tasa de Supervivencia , Células TH1/citología , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/citología , Células Th2/metabolismo , Transcriptoma , Trasplante Homólogo
9.
Xenotransplantation ; 21(6): 567-73, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25041432

RESUMEN

For the sake of therapy of diabetes, it is critical to understand human beta cell function in detail in health and disease. Current studies of human beta cell physiology in vivo are mostly limited to immunodeficient mouse models, which possess significant technical limitations. This study aimed to create a new model for the study of human islets through induction of transplant tolerance in immunosufficient mice. B6 diabetic mice were transplanted with human islets and treated with anti-CD45RB. To assess whether anti-CD45RB-induced transplant tolerance requires B cells, B6 recipients received additional anti-CD20 or B6µMT-/- mice were used. For some anti-CD45RB-treated B6µMT-/- mice, additional anti-CD25 mAb was applied at the early or late stage post-transplant. Immunohistology was performed to show the Foxp3 cells in grafted anti-CD45RB/anti-CD20-treated Foxp3-GFP B6 mice. The results showed that anti-CD45RB alone allowed indefinite graft survival in 26.6% of B6 mice, however 100% of xenografts were accepted in mice treated simultaneously with anti-CD20, and 88.9% of xenografts accepted in anti-CD45RB-treated µMT-/- mice. These µMT-/- mice accepted the islets from another human donor but rejected the islets from baboon. Additional administration of anti-CD25 mAb at the time of transplantation resulted in 100% rejection, whereas 40% of grafts were rejected while the antibody was administrated at days 60 post-transplant. Immunohistologic examination showed Foxp3+ cells accumulated around grafts. We conclude that induction of tolerance to human islets in an immunosufficient mouse model could be generated by targeting murine CD45RB and CD20. This new system will facilitate study of human islets and accelerate the dissection of the critical mechanisms underlying islet health in human disease.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Tolerancia al Trasplante/inmunología , Trasplante Heterólogo , Animales , Anticuerpos/inmunología , Diabetes Mellitus Experimental/cirugía , Rechazo de Injerto/inmunología , Humanos , Ratones Endogámicos C57BL , Trasplante Heterólogo/métodos
10.
Eur J Immunol ; 44(6): 1728-36, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24700192

RESUMEN

Regulatory B (Breg) cells have been shown to play a critical role in immune homeostasis and in autoimmunity models. We have recently demonstrated that combined anti-T cell immunoglobulin domain and mucin domain-1 and anti-CD45RB antibody treatment results in tolerance to full MHC-mismatched islet allografts in mice by generating Breg cells that are necessary for tolerance. Breg cells are antigen-specific and are capable of transferring tolerance to untreated, transplanted animals. Here, we demonstrate that adoptively transferred Breg cells require the presence of regulatory T (Treg) cells to establish tolerance, and that adoptive transfer of Breg cells increases the number of Treg cells. Interaction with Breg cells in vivo induces significantly more Foxp3 expression in CD4(+) CD25(-) T cells than with naive B cells. We also show that Breg cells express the TGF-ß associated latency-associated peptide and that Breg-cell mediated graft prolongation post-adoptive transfer is abrogated by neutralization of TGF-ß activity. Breg cells, like Treg cells, demonstrate preferential expression of both C-C chemokine receptor 6 and CXCR3. Collectively, these findings suggest that in this model of antibody-induced transplantation tolerance, Breg cells promote graft survival by promoting Treg-cell development, possibly via TGF-ß production.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/inmunología , Tolerancia al Trasplante , Traslado Adoptivo , Aloinjertos , Animales , Subgrupos de Linfocitos B/patología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Supervivencia de Injerto/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores CCR6/genética , Receptores CCR6/inmunología , Receptores CXCR3/genética , Receptores CXCR3/inmunología , Linfocitos T Reguladores/patología , Factor de Crecimiento Transformador beta/genética
11.
Cell Transplant ; 23(1): 51-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23192154

RESUMEN

A short course of anti-CD45RB leads to long-term islet allograft survival and donor-specific tolerance in approximately half of immunocompetent mice. We have previously demonstrated that anti-CD45RB antibody-mediated tolerance requires B-cells for cardiac allograft survival. We therefore asked whether B-cells were also required for anti-CD45RB antibody-mediated survival of islets. Unexpectedly, we found that nearly 100% of islet allografts survive long term in B-cell-deficient mice. Similarly, B-cell depletion by anti-CD22/cal augmented anti-CD45RB-mediated tolerance when administered pretransplant, although it had no effect on tolerance induction when administered posttransplant. Our results demonstrate that the role of B-cells in promoting tolerance with anti-CD45RB is graft specific, promoting tolerance in cardiac grafts but resisting tolerance in islet transplantation. These findings may help elucidate the varied action of B-cells in promoting tolerance versus rejection.


Asunto(s)
Linfocitos B/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/métodos , Tolerancia al Trasplante/inmunología , Aloinjertos , Animales , Linfocitos B/citología , Diabetes Mellitus Experimental/cirugía , Modelos Animales de Enfermedad , Antígenos Comunes de Leucocito/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos
12.
PLoS One ; 8(12): e82856, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24340063

RESUMEN

Immunosenescence predisposes the elderly to infectious and autoimmune diseases and impairs the response to vaccination. We recently demonstrated that ageing also impedes development of transplantation tolerance. Unlike their young counterparts (8-12 weeks of age) aged male recipients (greater than 12 months of age) transplanted with a full MHC-mismatched heart are resistant to tolerance mediated by anti-CD45RB antibody. Surprisingly, either chemical or surgical castration restored tolerance induction to levels observed using young recipients. Based on the strong impact of endocrine modulation on transplant tolerance, we explored the impact of ageing and castration on the immune system. Here we report a significant increase in the percentage of T cells that produce interferon-γ (IFN-γ) in aged male versus young male animals and that the overall increase in IFN-γ production was due to an expansion of IFN-γ-producing memory T cells in aged animals. In contrast to IFN-γ production, we did not observe differences in IL-10 expression in young versus old male mice. We hypothesized that endocrine modulation would diminish the elevated levels of IFN-γ production in aged recipients, however, we observed no significant reduction in the percentage of IFN-γ+ T cells upon castration. Furthermore, we neutralized interferon-γ by antibody and did not observe an effect on graft survival. We conclude that while elevated levels of interferon-γ serves as a marker of tolerance resistance in aged mice, other as yet to be identified factors are responsible for its cause. Defining these factors may be relevant to design of tolerogenic strategies for aged recipients.


Asunto(s)
Memoria Inmunológica/fisiología , Interferón gamma/biosíntesis , Linfocitos T/citología , Tolerancia al Trasplante/inmunología , Factores de Edad , Animales , Rechazo de Injerto , Supervivencia de Injerto , Interleucina-10/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Modelos Animales , Trasplante de Piel , Linfocitos T/inmunología
13.
Transplant Rev (Orlando) ; 27(2): 61-4, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23474287

RESUMEN

One of the most significant advances in the field of immunology in the last decade is delineation of the pivotal role of regulatory T cells (Tregs) in the maintenance of self-tolerance. While Tregs are just now being applied therapeutically in early phase clinical trials, data gleaned from basic and translational studies to-date suggest enormous potential to intervene in human disease. Data from our work and the work of others suggest that the innate immune system plays an important role in the differentiation and function of Tregs, largely through the production of cytokines but also through expression of cell surface ligands. These molecules are expressed differentially depending on whether the stimulus includes trauma, ischemia/necrosis, and microbial infection, and have opposing effects on Tregs, in contrast to those associated with dendritic cell maturation and somatic cell apoptosis, which promote Treg differentiation and function. We refer to the former process as Treg counter-regulation. Since the transplantation procedure involves surgical trauma, organ ischemia, and exposure to environmental microbes, Treg counter-regulation represents a key area of intervention to improve strategies for promoting allograft tolerance.


Asunto(s)
Tolerancia Inmunológica/inmunología , Inmunidad Innata/inmunología , Linfocitos T Reguladores/inmunología , Inmunología del Trasplante , Humanos
14.
Nat Biotechnol ; 29(11): 1005-10, 2011 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-21983520

RESUMEN

Excessive and prolonged activity of inflammatory monocytes is a hallmark of many diseases with an inflammatory component. In such conditions, precise targeting of these cells could be therapeutically beneficial while sparing many essential functions of the innate immune system, thus limiting unwanted effects. Inflammatory monocytes-but not the noninflammatory subset-depend on the chemokine receptor CCR2 for localization to injured tissue. Here we present an optimized lipid nanoparticle and a CCR2-silencing short interfering RNA that, when administered systemically in mice, show rapid blood clearance, accumulate in spleen and bone marrow, and localize to monocytes. Efficient degradation of CCR2 mRNA in monocytes prevents their accumulation in sites of inflammation. Specifically, the treatment attenuates their number in atherosclerotic plaques, reduces infarct size after coronary artery occlusion, prolongs normoglycemia in diabetic mice after pancreatic islet transplantation, and results in reduced tumor volumes and lower numbers of tumor-associated macrophages.


Asunto(s)
Silenciador del Gen , Inflamación/terapia , Macrófagos/efectos de los fármacos , Nanopartículas , ARN Interferente Pequeño/uso terapéutico , Receptores CCR2/antagonistas & inhibidores , Animales , Aterosclerosis/terapia , Glucemia , Diabetes Mellitus/cirugía , Diabetes Mellitus/terapia , Modelos Animales de Enfermedad , Supervivencia de Injerto/genética , Humanos , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Infarto del Miocardio/prevención & control , Infarto del Miocardio/terapia , Nanopartículas/química , Receptores CCR2/genética
15.
Sci Transl Med ; 3(87): 87ra52, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21677198

RESUMEN

The senescent immune system responds poorly to new stimuli; thymic involution, accumulation of memory cells against other specificities, and general refractoriness to antigen signaling all may contribute to poor resistance to infection. These same changes may pose a significant clinical barrier to organ transplantation, as transplantation tolerance requires thymic participation and integrated, tolerance-promoting responses to novel antigens. We found that after the age of 12 months, mice became resistant to the tolerance-inducing capacity of the monoclonal antibody therapy anti-CD45RB. This resistance to tolerance to cardiac allografts could be overcome by surgical castration of male mice, a procedure that led to thymic regeneration and long-term graft acceptance. The potential for clinical translation of this endocrine-immune interplay was confirmed by the ability of Lupron Depot injections, which temporarily disrupt gonadal function, to restore tolerance in aged mice. Furthermore, we demonstrated that the restoration of tolerance after surgical or chemical castration depended on thymic production of regulatory T cells (T(regs)); thymectomy or T(reg) depletion abrogated tolerance restoration. The aging of the immune system ("immune senescence") is a significant barrier to immune tolerance, but this barrier can be overcome by targeting sex steroid production with commonly used clinical therapeutics.


Asunto(s)
Envejecimiento/inmunología , Sistema Endocrino/fisiología , Sistema Inmunológico/fisiología , Tolerancia al Trasplante/inmunología , Animales , Antineoplásicos Hormonales/farmacología , Castración , Gónadas/efectos de los fármacos , Gónadas/fisiología , Trasplante de Corazón/inmunología , Humanos , Antígenos Comunes de Leucocito/inmunología , Leuprolida/farmacología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Timectomía , Timo/citología , Timo/efectos de los fármacos , Trasplante Homólogo/inmunología
16.
Transplantation ; 91(7): 707-13, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21386770

RESUMEN

BACKGROUND: Because CD4CD25Foxp3 regulatory T cells (Tregs) are essential for the maintenance of self-tolerance, significant interest surrounds the developmental cues for thymic-derived natural Tregs (nTregs) and periphery-generated adaptive Tregs (aTregs). In the transplant setting, the allograft may play a role in the generation of alloantigen-specific Tregs, but this role remains undefined. We examined whether the immune response to a transplant allograft results in the peripheral generation of aTregs. METHODS: To identify generation of aTregs, purified graft-reactive CD4CD25 T cells were adoptively transferred to mice-bearing skin allograft. To demonstrate that aTregs are necessary for tolerance, DBA/2 skin was transplanted onto C57BL/6-RAG-1-deficient recipients adoptively transferred with purified sorted CD4CD25 T cells; half of the recipients undergo tolerance induction treatment. RESULTS: By tracking adoptively transferred cells, we show that purified graft-reactive CD4CD25 T lymphocytes up-regulate Foxp3 in mice receiving skin allografts in the absence of any treatment. Interestingly, cotransfer of antigen-specific nTregs suppresses the up-regulation of Foxp3 by inhibiting the proliferation of allograft-responsive T cells. In vitro data are consistent with our in vivo data-Foxp3 cells are generated on antigen activation, and this generation is suppressed on coculture with antigen-specific nTregs. Finally, blocking aTreg generation in grafted, rapamycin-treated mice disrupts alloantigen-specific tolerance induction. In contrast, blocking aTreg generation in grafted mice treated with nondepleting anti-CD4 plus anti-CD40L antibodies does not disrupt graft tolerance. CONCLUSIONS: We conclude that graft alloantigen stimulates the de novo generation of aTregs, and this generation may represent a necessary step in some but not all protocols of tolerance induction.


Asunto(s)
Isoantígenos/inmunología , Linfocitos T Reguladores/fisiología , Tolerancia al Trasplante , Animales , Antígenos CD40/fisiología , Ligando de CD40/fisiología , Factores de Transcripción Forkhead/genética , Interleucina-2/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Sirolimus/farmacología , Factor de Crecimiento Transformador beta/farmacología , Trasplante Homólogo
17.
Eur J Immunol ; 40(5): 1369-74, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20148423

RESUMEN

Involvement of Treg in transplant tolerance has been demonstrated in multiple models. During the active process of graft rejection, these regulatory cells are themselves regulated and inactivated, a process termed counter-regulation. We hypothesize that ligation of the costimulatory molecule glucocorticoid-induced TNF receptor-related protein (GITR) on Treg inhibits their ability to promote graft survival, and by blocking GITR ligation graft survival can be prolonged. To this aim, we have designed a soluble GITR fusion protein (GITR-Fc), which binds GITR ligand and inhibits activation of GITR. Here, we show that GITR-Fc prolonged mouse skin graft survival, and this prolongation is dependent on Treg. In a full MHC-mismatched skin graft setting, GITR-Fc significantly improved graft survival when used in combination with MR1, anti-CD40L, while GITR-Fc alone did not demonstrate graft prolongation. These results demonstrate that disruption of binding of GITR with GITR ligand may be an important strategy in prolonging allograft survival.


Asunto(s)
Supervivencia de Injerto/efectos de los fármacos , Inmunosupresores/uso terapéutico , Receptores de Factor de Crecimiento Nervioso/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Trasplante de Piel/inmunología , Linfocitos T Reguladores/inmunología , Trasplante Homólogo/inmunología , Inhibidores del Factor de Necrosis Tumoral , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Unión Competitiva , Ligando de CD40 , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Antígenos de Histocompatibilidad Clase I , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Antígenos de Histocompatibilidad Menor , Receptores de Factor de Crecimiento Nervioso/genética , Receptores del Factor de Necrosis Tumoral/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Factores de Necrosis Tumoral/inmunología
18.
Transplantation ; 88(6): 820-5, 2009 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-19920782

RESUMEN

BACKGROUND: Isolated islet transplantation with infusions from two to three donor pancreata and Edmonton immunosuppression consistently achieves insulin independence in patients with type 1 diabetes. The success of this protocol has been attributed to a novel combination of immunosuppressive agents and avoidance of steroids; however, the outcome of islet transplantation may differ in kidney transplant recipients who are already immunosuppressed. METHODS: We compared the metabolic outcomes and graft survival of islet transplantation in our program where nine patients underwent islet transplantation alone treated with Edmonton immunosuppression and eight patients received islet after kidney (IAK) transplants under standard kidney transplant immunosuppression often including steroids. RESULTS: Transplants in the IAK and islet transplantation alone setting demonstrated similar islet potency (islet equivalents/unit insulin reduction) and recipients from both groups routinely gained insulin independence, functional islet mass, and duration of graft survival, however, seemed superior in the IAK group. CONCLUSIONS: These results suggest that better islet graft function and survival may be attained using non-Edmonton rather than Edmonton immunosuppression and can include maintenance steroid therapy.


Asunto(s)
Trasplante de Islotes Pancreáticos/métodos , Trasplante de Islotes Pancreáticos/fisiología , Trasplante de Riñón/métodos , Adulto , Glucemia/metabolismo , Péptido C/sangre , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/cirugía , Femenino , Hemoglobina Glucada/metabolismo , Supervivencia de Injerto/fisiología , Humanos , Terapia de Inmunosupresión/métodos , Insulina/administración & dosificación , Masculino , Persona de Mediana Edad , Factores de Tiempo
19.
Transplantation ; 88(10): 1169-77, 2009 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-19935370

RESUMEN

BACKGROUND: Many models of transplant tolerance have been found to depend on the induction of regulatory T cells (Tregs). Innate immune signals are known to suppress Tregs thereby augmenting immunity by abrogating Treg function. Such signals may also provide a barrier to transplantation tolerance mediated by Tregs. A number of cell surface molecules expressed by Tregs have been found to inhibit Treg activity, the best characterized of which is the glucocorticoid-induced tumor necrosis factor receptor-related (GITR) protein. METHODS: By using an adoptive transfer model of allograft rejection, we can study the effects of inflammation and antigen-specific Tregs on graft survival. Inflammation resulting from the transplant procedure counter-regulates the suppressor activity of Tregs. To assess whether Treg activity could be enhanced by blocking GITR signaling, we compared the capacity of Tregs to prolong the survival of grafts in the presence or absence of activation-inducible TNF receptor (AITRL)-Fc, a novel construct that binds GITR. RESULTS: We report that interruption of GITR-GITR ligand (GITRL) binding by AITRL-Fc resulted in long-term Treg-dependent acceptance of skin grafts in the setting of innate immune signals that otherwise interfere with Treg activity. CONCLUSIONS: Inflammation and other innate immune signals may activate antigen presenting cells to upregulate GITRL. GITR-GITRL interaction is one pathway by which antigen presenting cells may enhance the adaptive response to foreign antigen by counter-regulating Tregs and by costimulating effector T cells. By blocking this interaction with AITRL-Fc, one can sustain the benefit conferred by graft-protective Tregs.


Asunto(s)
Supervivencia de Injerto/inmunología , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Trasplante de Piel/inmunología , Linfocitos T Reguladores/inmunología , Trasplante Homólogo/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Cruzamientos Genéticos , Epítopos/inmunología , Citometría de Flujo , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Humanos , Terapia de Inmunosupresión , Inflamación/inmunología , Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Receptores de Factor de Crecimiento Nervioso/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal/inmunología , Transducción de Señal/fisiología , Trasplante de Piel/mortalidad , Tasa de Supervivencia , Factores de Necrosis Tumoral/inmunología
20.
Transplantation ; 85(5): 675-80, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18337659

RESUMEN

BACKGROUND: Allogeneic tolerance can be reliably obtained with monoclonal antibody therapy targeting CD45RB. Although regulatory T cells play an important role in the mechanism, we have recently demonstrated the active participation of host B lymphocytes. After anti-CD45RB therapy, B lymphocytes demonstrate phenotypic alterations that include up-regulation of CD54 (intercellular adhesion molecule [ICAM]-1). We have investigated the hypothesis that alteration in ICAM-1 expression is required for tolerance induction. MATERIALS AND METHODS: Recipients of heterotopic allogeneic cardiac grafts (C3H donors into B6 recipients) were treated with anti-CD45RB, anti-ICAM, anti-lymphocyte function-associated antigen-1 (LFA), or the combination of these agents. These data were extended by performing allogeneic cardiac transplants into ICAM or LFA recipients treated with a 5-day course of anti-CD45RB. Finally, B-cell-deficient animals were reconstituted with ICAM splenocytes to create a recipient with a selective deficiency of ICAM-1 restricted to the B-cell compartment. RESULTS: Anti-CD45RB alone or the combination of anti-LFA/anti-ICAM reliably induced transplantation tolerance. However, the triple combination was routinely unsuccessful and induced long-term graft survival in no recipients. ICAM-deficient or LFA-deficient recipients were also resistant to tolerance induced by anti-CD45RB. Finally, transfer of control splenocytes to B-cell-deficient recipients permitted anti-CD45RB-induced tolerance, whereas transfer of ICAM cells was unable to support tolerance induction. CONCLUSIONS: Expression of ICAM-1 by B lymphocytes and interaction with LFA-1 form a central aspect of transplantation tolerance induced by anti-CD45RB therapy. These data further elucidate the cellular mechanisms used by B lymphocytes in the induction of transplantation tolerance.


Asunto(s)
Linfocitos B/inmunología , Trasplante de Corazón/inmunología , Molécula 1 de Adhesión Intercelular/fisiología , Antígenos Comunes de Leucocito/inmunología , Antígeno-1 Asociado a Función de Linfocito/fisiología , Linfocitos T/inmunología , Tolerancia al Trasplante/inmunología , Animales , Anticuerpos/inmunología , Citometría de Flujo , Molécula 1 de Adhesión Intercelular/genética , Antígenos Comunes de Leucocito/genética , Antígeno-1 Asociado a Función de Linfocito/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/inmunología , Trasplante Homólogo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...