Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
J Am Coll Cardiol ; 74(1): 36-51, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31272550

RESUMO

BACKGROUND: The CTOT-11 (Prevention of Cardiac Allograft Vasculopathy Using Rituximab Therapy in Cardiac Transplantation [Clinical Trials in Organ Transplantation-11]) study was a randomized, placebo-controlled, multicenter, double-blinded clinical trial in nonsensitized primary heart transplant (HTX) recipients. OBJECTIVES: The study sought to determine whether B cell depletion therapy would attenuate the development of cardiac allograft vasculopathy. METHODS: A total of 163 HTX recipients were randomized to rituximab 1,000 mg intravenous or placebo on days 0 and 12 post-transplant. Primary outcome was change in percent atheroma volume (PAV) from baseline to 1 year measured by intravascular ultrasound. Secondary outcomes included treated episodes of acute rejection, de novo anti-HLA antibodies (including donor-specific antibodies), and phenotypic differentiation of B cells. RESULTS: There were no significant differences at study entry between the rituximab and placebo groups. Paired intravascular ultrasound measures were available at baseline and 1 year in 86 subjects (49 rituximab, 37 placebo). The mean ± SD change in PAV at 12 months was +6.8 ± 8.2% rituximab versus +1.9 ± 4.4% placebo (p = 0.0019). Mortality at 12 months was 3.4% rituximab versus 6.8% placebo (p = 0.47); there were no retransplants or post-transplant lymphoproliferative disorder. The rate of treated rejection was 24.7% rituximab versus 32.4% placebo (p = 0.28). Rituximab therapy effectively eliminated CD20+/CD19+ B cells followed by a gradual expansion of a CD19- cell population in the rituximab-treated group. CONCLUSIONS: A marked, unexpected increase in coronary artery PAV with rituximab was observed during the first year in HTX recipients. One-year mortality was not impacted; however, longer-term follow-up and mechanistic explanations are required. (Prevention of Cardiac Allograft Vasculopathy Using Rituximab [Rituxan] Therapy in Cardiac Transplantation; NCT01278745).


Assuntos
Transplante de Coração , Fatores Imunológicos/uso terapêutico , Complicações Pós-Operatórias/prevenção & controle , Rituximab/uso terapêutico , Doenças Vasculares/prevenção & controle , Adulto , Aloenxertos , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Adulto Jovem
2.
Am J Pathol ; 187(6): 1368-1379, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28427861

RESUMO

Costimulatory molecules, such as the programmed death ligand (PD-L1), might exert differential effects on T-cell function, depending on the clinical setting and/or immunological environment. Given the impact of T cells on bronchiolitis obliterans (BO) in lung transplantation, we used an established tracheal transplant model inducing BO-like lesions to investigate the impact of PD-L1 on alloimmune responses and histopathological outcome in BO. In contrast to other transplant models in which PD-L1 generally shows protective functions, we demonstrated that PD-L1 has divergent effects depending on its location in donor versus recipient tissue. Although PD-L1 deficiency in donor tissue worsened histopathological outcome, and increased systemic inflammatory response, recipient PD-L1 deficiency induced opposite effects. Mechanistic studies revealed PD-L1-deficient recipients were hyporesponsive toward alloantigen, despite increased numbers of CD8+ effector T cells. The function of PD-L1 on T cells after unspecific stimulation was dependent on both cell type and strength of stimulation. This novel function of recipient PD-L1 may result from the high degree of T-cell activation within the highly immunogenic milieu of the transplanted tissue. In this model, both decreased T-cell alloimmune responses and the reduction of BO in PD-L1-deficient recipients suggest a potential therapeutic role of selectively blocking PD-L1 in the recipient. Further investigation is warranted to determine the impact of this finding embedded in the complex pathophysiological context of BO.


Assuntos
Antígeno B7-H1/imunologia , Bronquiolite Obliterante/imunologia , Traqueia/transplante , Imunologia de Transplantes , Animais , Antígeno B7-H1/deficiência , Bronquiolite Obliterante/patologia , Bronquiolite Obliterante/prevenção & controle , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Células Epiteliais/imunologia , Sobrevivência de Enxerto/imunologia , Tolerância Imunológica/imunologia , Imunidade Celular , Isoantígenos/imunologia , Ativação Linfocitária/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Doadores de Tecidos , Traqueia/patologia , Regulação para Cima/imunologia
3.
Diabetes ; 64(1): 158-71, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25187361

RESUMO

Impaired regulatory B cell (Breg) responses are associated with several autoimmune diseases in humans; however, the role of Bregs in type 1 diabetes (T1D) remains unclear. We hypothesized that naturally occurring, interleukin-10 (IL-10)-producing Bregs maintain tolerance to islet autoantigens, and that hyperglycemic nonobese diabetic (NOD) mice and T1D patients lack these potent negative regulators. IgVH transcriptome analysis revealed that islet-infiltrating B cells in long-term normoglycemic (Lnglc) NOD, which are naturally protected from diabetes, are more antigen-experienced and possess more diverse B-cell receptor repertoires compared to those of hyperglycemic (Hglc) mice. Importantly, increased levels of Breg-promoting CD40(+) B cells and IL-10-producing B cells were found within islets of Lnglc compared to Hglc NOD. Likewise, healthy individuals showed increased frequencies of both CD40(+) and IL-10(+) B cells compared to T1D patients. Rituximab-mediated B-cell depletion followed by adoptive transfer of B cells from Hglc mice induced hyperglycemia in Lnglc human CD20 transgenic NOD mouse models. Importantly, both murine and human IL-10(+) B cells significantly abrogated T-cell-mediated responses to self- or islet-specific peptides ex vivo. Together, our data suggest that antigen-matured Bregs may maintain tolerance to islet autoantigens by selectively suppressing autoreactive T-cell responses, and that Hglc mice and individuals with T1D lack this population of Bregs.


Assuntos
Linfócitos B Reguladores/imunologia , Antígenos CD40/imunologia , Diabetes Mellitus Tipo 1/imunologia , Tolerância Imunológica/imunologia , Interleucina-10/imunologia , Ilhotas Pancreáticas/imunologia , Adulto , Animais , Autoantígenos/genética , Autoantígenos/imunologia , Linfócitos B Reguladores/metabolismo , Antígenos CD40/metabolismo , Feminino , Humanos , Interleucina-10/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Pessoa de Meia-Idade , Transcriptoma/imunologia , Adulto Jovem
4.
PLoS One ; 9(2): e89561, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586872

RESUMO

AIMS/HYPOTHESIS: Inhibition of PD1-PDL1 signaling in NOD mice accelerates onset of type 1 diabetes implicating this pathway in suppressing the emergence of pancreatic beta cell reactive T-cells. However, the molecular mechanism by which PD1 signaling protects from type 1 diabetes is not clear. We hypothesized that differential susceptibility of Idd mouse strains to type 1 diabetes when challenged with anti PDL1 will identify genomic loci that collaborate with PD1 signaling in suppressing type 1 diabetes. METHODS: Anti PDL1 was administered to NOD and various Idd mouse strains at 10 weeks of age and onset of disease was monitored by measuring blood glucose levels. Additionally, histological evaluation of the pancreas was performed to determine degree of insulitis. Statistical analysis of the data was performed using Log-Rank and Student's t-test. RESULTS: Blockade of PDL1 rapidly precipitated type 1 diabetes in nearly all NOD Idd congenic strains tested, despite the fact that all are moderately (Idd5, Idd3 and Idd10/18) or highly (Idd3/10/18 and Idd9) protected from spontaneous type 1 diabetes by virtue of their protective Idd genes. Only the Idd3/5 strain, which is nearly 100% protected from spontaneous disease, remained normoglycemic following PDL1 blockade. CONCLUSIONS: These results indicate that multiple Idd loci collaborate with PD1 signaling. Anti PDL1 treatment undermines a large portion of the genetic protection mediated by Idd genes in the NOD model of type 1 diabetes. Basal insulitis correlated with higher susceptibility to type 1 diabetes. These findings have important implications since the PD1 pathway is a target for immunotherapy.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Transferência Adotiva , Animais , Antígeno B7-H1/metabolismo , Diabetes Mellitus Tipo 1/genética , Feminino , Predisposição Genética para Doença , Imunoterapia , Camundongos , Camundongos Endogâmicos NOD , Receptor de Morte Celular Programada 1 , Transdução de Sinais
5.
J Am Soc Nephrol ; 25(7): 1415-29, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24676639

RESUMO

Podocyte injury and resulting albuminuria are hallmarks of diabetic nephropathy, but targeted therapies to halt or prevent these complications are currently not available. Here, we show that the immune-related molecule B7-1/CD80 is a critical mediator of podocyte injury in type 2 diabetic nephropathy. We report the induction of podocyte B7-1 in kidney biopsy specimens from patients with type 2 diabetes. Genetic and epidemiologic studies revealed the association of two single nucleotide polymorphisms at the B7-1 gene with diabetic nephropathy. Furthermore, increased levels of the soluble isoform of the B7-1 ligand CD28 correlated with the progression to ESRD in individuals with type 2 diabetes. In vitro, high glucose conditions prompted the phosphatidylinositol 3 kinase-dependent upregulation of B7-1 in podocytes, and the ectopic expression of B7-1 in podocytes increased apoptosis and induced disruption of the cytoskeleton that were reversed by the B7-1 inhibitor CTLA4-Ig. Podocyte expression of B7-1 was also induced in vivo in two murine models of diabetic nephropathy, and treatment with CTLA4-Ig prevented increased urinary albumin excretion and improved kidney pathology in these animals. Taken together, these results identify B7-1 inhibition as a potential therapeutic strategy for the prevention or treatment of diabetic nephropathy.


Assuntos
Antígeno B7-1/fisiologia , Diabetes Mellitus Tipo 1/complicações , Nefropatias Diabéticas/etiologia , Podócitos , Adulto , Idoso , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Regulação para Cima
6.
Expert Opin Ther Targets ; 18(2): 225-42, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24329604

RESUMO

INTRODUCTION: The pivotal role of costimulatory pathways in regulating T-cell activation versus tolerance has stimulated tremendous interest in their manipulation for therapeutic purposes. Of these, the CD28-B7 pathway is arguably the most important and best studied. Therapeutic targets of CD28 are currently used in the treatment of melanoma, autoimmune diseases and in transplantation. AREAS COVERED: In this review, we summarize our current knowledge of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) signaling, and review the current state and challenges of harnessing them to promote transplant tolerance. EXPERT OPINION: Despite the success of belatacept, a first-in-class CTLA-4 fusion protein now clinically used in transplantation, it is apparent that we have only scratched the surface in understanding the complexities of how costimulatory pathways modulate the immune system. Our initial assumption that positive costimulators activate effector T cells and prevent tolerance, while negative costimulators inhibit effector T cells and promote tolerance, is clearly an oversimplified view. Indeed, belatacept is not only capable of blocking deleterious CD28-B7 interactions that promote effector T-cell responses but can also have undesired effects on tolerogenic regulatory T-cell populations.


Assuntos
Antígenos CD28/imunologia , Rejeição de Enxerto/prevenção & controle , Animais , Antígeno CTLA-4/imunologia , Humanos , Linfócitos T/imunologia
7.
Expert Opin Biol Ther ; 13(11): 1557-68, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24083381

RESUMO

INTRODUCTION: The concern about nephrotoxicity with calcineurin inhibitors led to the search of novel agents for immunosuppression. Based on the requirement of T-cell co-stimulatory signals to fully activated naïve T cells, it became clear that blocking these pathways could be an appealing therapeutic target. However, some unexpected findings were noticed in the recent clinical trials of belatacept, including a higher rate of rejection, which warranted further investigation with some interesting concepts emerging from the bench. AREAS COVERED: This article aims to review the literature of the B7:CD28 co-stimulatory blockade in transplantation, including the basic immunology behind its development, clinical application and potential limitations. EXPERT OPINION: Targeting co-stimulatory pathways were found to be much more complex than initially anticipated due to the interplay between not only various co-stimulatory pathways but also various co-inhibitory ones. In addition, co-stimulatory signals have different roles in diverse immune cell types. Therefore, targeting CD28 ligands with cytotoxic T lymphocyte antigen-4 (CTLA4)-Ig may have some deleterious effects, including the inhibition of regulatory T cells, blockade of co-inhibitory signals (CTLA4) and promotion of Th17 cells. Co-stimulatory independence of memory T cells was another unforeseen limitation. Learning how to better integrate co-stimulatory targeting with other immunosuppressive agents will be critical for the improvement of long-term graft survival.


Assuntos
Antígenos CD28/antagonistas & inibidores , Receptores Coestimuladores e Inibidores de Linfócitos T/efeitos dos fármacos , Imunossupressores/uso terapêutico , Linfócitos T Citotóxicos/efeitos dos fármacos , Imunologia de Transplantes , Abatacepte , Animais , Antígeno B7-1 , Antígeno B7-2/antagonistas & inibidores , Antígeno B7-2/imunologia , Antígenos CD28/agonistas , Antígenos CD28/imunologia , Ensaios Clínicos como Assunto , Ciclosporina/efeitos adversos , Ciclosporina/uso terapêutico , Suscetibilidade a Doenças , Facilitação Imunológica de Enxerto/métodos , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Imunoconjugados/efeitos adversos , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Memória Imunológica/efeitos dos fármacos , Imunossupressores/efeitos adversos , Imunossupressores/farmacologia , Infecções/etiologia , Nefropatias/induzido quimicamente , Nefropatias/prevenção & controle , Ativação Linfocitária/efeitos dos fármacos , Transtornos Linfoproliferativos/induzido quimicamente , Terapia de Alvo Molecular , Estudos Multicêntricos como Assunto , Complicações Pós-Operatórias/induzido quimicamente , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia
8.
PLoS One ; 8(4): e60391, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593209

RESUMO

CD160 is a cell surface molecule expressed by most NK cells and approximately 50% of CD8(+) cytotoxic T lymphocytes. Engagement of CD160 by MHC class-I directly triggers a costimulatory signal to TCR-induced proliferation, cytokine production and cytotoxic effector functions. The role of CD160 in alloimmunity is unknown. Using a newly generated CD160 fusion protein (CD160Ig) we examined the role of the novel costimulatory molecule CD160 in mediating CD4(+) or CD8(+) T cell driven allograft rejection. CD160Ig inhibits alloreactive CD8(+) T cell proliferation and IFN-γ production in vitro, in particular in the absence of CD28 costimulation. Consequently CD160Ig prolongs fully mismatched cardiac allograft survival in CD4(-/-), CD28(-/-) knockout and CTLA4Ig treated WT recipients, but not in WT or CD8(-/-) knockout recipients. The prolonged cardiac allograft survival is associated with reduced alloreactive CD8(+) T cell proliferation, effector/memory responses and alloreactive IFN-γ production. Thus, CD160 signaling is particularly important in CD28-independent effector/memory CD8(+) alloreactive T cell activation in vivo and therefore may serve as a novel target for prevention of allograft rejection.


Assuntos
Antígenos CD/imunologia , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/imunologia , Imunoglobulina G/imunologia , Receptores Imunológicos/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD/genética , Antígenos CD28/deficiência , Antígenos CD28/imunologia , Antígenos CD4/genética , Antígenos CD4/imunologia , Citocinas/biossíntese , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Expressão Gênica , Sobrevivência de Enxerto/genética , Transplante de Coração/imunologia , Transplante de Coração/mortalidade , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Imunoglobulina G/genética , Memória Imunológica/genética , Memória Imunológica/imunologia , Interferon gama/biossíntese , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Imunológicos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Transplante de Pele/imunologia , Transplante de Pele/mortalidade , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Transplante Homólogo
9.
Diabetes ; 62(5): 1665-75, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23315496

RESUMO

The lymphocytic ionotropic purinergic P2X receptors (P2X1R-P2X7R, or P2XRs) sense ATP released during cell damage-activation, thus regulating T-cell activation. We aim to define the role of P2XRs during islet allograft rejection and to establish a novel anti-P2XRs strategy to achieve long-term islet allograft function. Our data demonstrate that P2X1R and P2X7R are induced in islet allograft-infiltrating cells, that only P2X7R is increasingly expressed during alloimmune response, and that P2X1R is augmented in both allogeneic and syngeneic transplantation. In vivo short-term P2X7R targeting (using periodate-oxidized ATP [oATP]) delays islet allograft rejection, reduces the frequency of Th1/Th17 cells, and induces hyporesponsiveness toward donor antigens. oATP-treated mice displayed preserved islet grafts with reduced Th1 transcripts. P2X7R targeting and rapamycin synergized in inducing long-term islet function in 80% of transplanted mice and resulted in reshaping of the recipient immune system. In vitro P2X7R targeting using oATP reduced T-cell activation and diminished Th1/Th17 cytokine production. Peripheral blood mononuclear cells obtained from long-term islet-transplanted patients showed an increased percentage of P2X7R⁺CD4⁺ T cells compared with controls. The beneficial effects of oATP treatment revealed a role for the purinergic system in islet allograft rejection, and the targeting of P2X7R is a novel strategy to induce long-term islet allograft function.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Rejeição de Enxerto/prevenção & controle , Terapia de Imunossupressão , Transplante das Ilhotas Pancreáticas/efeitos adversos , Antagonistas do Receptor Purinérgico P2X/uso terapêutico , Receptores Purinérgicos P2X7/metabolismo , Transplante Heterotópico/efeitos adversos , Trifosfato de Adenosina/uso terapêutico , Adulto , Animais , Feminino , Humanos , Imunossupressores/uso terapêutico , Transplante das Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Receptores Purinérgicos P2X7/química , Receptores Purinérgicos P2X7/genética , Sirolimo/uso terapêutico , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Transplante Heterotópico/imunologia , Transplante Heterotópico/patologia , Transplante Homólogo , Transplante Isogênico
10.
Circulation ; 127(4): 463-75, 2013 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23250993

RESUMO

BACKGROUND: Heart transplantation is a lifesaving procedure for patients with end-stage heart failure. Despite much effort and advances in the field, current immunosuppressive regimens are still associated with poor long-term cardiac allograft outcomes, and with the development of complications, including infections and malignancies, as well. The development of a novel, short-term, and effective immunomodulatory protocol will thus be an important achievement. The purine ATP, released during cell damage/activation, is sensed by the ionotropic purinergic receptor P2X7 (P2X7R) on lymphocytes and regulates T-cell activation. Novel clinical-grade P2X7R inhibitors are available, rendering the targeting of P2X7R a potential therapy in cardiac transplantation. METHODS AND RESULTS: We analyzed P2X7R expression in patients and mice and P2X7R targeting in murine recipients in the context of cardiac transplantation. Our data demonstrate that P2X7R is specifically upregulated in graft-infiltrating lymphocytes in cardiac-transplanted humans and mice. Short-term P2X7R targeting with periodate-oxidized ATP promotes long-term cardiac transplant survival in 80% of murine recipients of a fully mismatched allograft. Long-term survival of cardiac transplants was associated with reduced T-cell activation, T-helper cell 1/T-helper cell 17 differentiation, and inhibition of STAT3 phosphorylation in T cells, thus leading to a reduced transplant infiltrate and coronaropathy. In vitro genetic upregulation of the P2X7R pathway was also shown to stimulate T-helper cell 1/T-helper cell 17 cell generation. Finally, P2X7R targeting halted the progression of coronaropathy in a murine model of chronic rejection as well. CONCLUSIONS: P2X7R targeting is a novel clinically relevant strategy to prolong cardiac transplant survival.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Rejeição de Enxerto/tratamento farmacológico , Rejeição de Enxerto/mortalidade , Transplante de Coração/mortalidade , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X7/metabolismo , Trifosfato de Adenosina/farmacologia , Adulto , Animais , Doença Crônica , Modelos Animais de Doenças , Feminino , Rejeição de Enxerto/imunologia , Transplante de Coração/imunologia , Humanos , Imunocompetência/efeitos dos fármacos , Imunocompetência/imunologia , Isoantígenos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/imunologia , Fator de Transcrição STAT3/metabolismo , Sobreviventes/estatística & dados numéricos , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia
11.
Eur J Immunol ; 42(9): 2343-53, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22733595

RESUMO

Ligands of the B7 family provide both positive and negative costimulatory signals to the CD28 family of receptors on T lymphocytes, the balance of which determines the immune response. B7-H3 is a member of the B7 family whose function in T-cell activation has been the subject of some controversy: in autoimmunity and tumor immunity, it has been described as both costimulatory and coinhibitory, while in transplantation, B7-H3 signaling is thought to contribute to graft rejection. However, we now demonstrate results to the contrary. Signaling through a putative B7-H3 receptor prolonged allograft survival in a fully MHC-mismatched cardiac model and promoted a shift toward a Th2 milieu; conversely, B7-H3 blockade, achieved by use of a blocking antibody, resulted in accelerated rejection, an effect associated with enhanced IFN-γ production. Finally, graft prolongation achieved by CTLA4 Ig was shortened both by B7-H3 blockade and the absence of recipient B7-H3. These findings suggest a coinhibitory role for B7-H3. However, experience with other CD28/B7 family members suggests that immune redundancy plays a crucial role in determining the functions of various pathways. Given the abundance of conflicting data, it is plausible that, under differing conditions, B7-H3 may have both positive and negative costimulatory functions.


Assuntos
Antígenos B7/imunologia , Transplante de Coração/imunologia , Transdução de Sinais/imunologia , Células Th1/imunologia , Transplante Homólogo/imunologia , Abatacepte , Animais , Antígenos B7/metabolismo , Antígenos CD28/imunologia , Antígenos CD28/metabolismo , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Imunoconjugados/imunologia , Imunoconjugados/metabolismo , Interferon gama/imunologia , Interferon gama/metabolismo , Cinética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Imunologia de Transplantes
12.
J Am Soc Nephrol ; 23(1): 174-82, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22052056

RESUMO

The incidence of developing circulating anti-human leukocyte antigen antibodies and the kinetics of T cell depletion and recovery among pediatric renal transplant recipients who receive alemtuzumab induction therapy are unknown. In a collaborative endeavor to minimize maintenance immunosuppression in pediatric renal transplant recipients, we enrolled 35 participants from four centers and treated them with alemtuzumab induction therapy and a steroid-free, calcineurin-inhibitor-withdrawal maintenance regimen. At 3 months after transplant, there was greater depletion of CD4(+) than CD8(+) T cells within the total, naive, memory, and effector memory subsets, although depletion of the central memory subset was similar for CD4(+) and CD8(+) cells. Although CD8(+) T cells recovered faster than CD4(+) subsets overall, they failed to return to pretransplant levels by 24 months after transplant. There was no evidence for greater recovery of either CD4(+) or CD8(+) memory cells than naïve cells. Alemtuzumab relatively spared CD4(+)CD25(+)FoxP3(+) regulatory T cells, resulting in a rise in their numbers relative to total CD4(+) cells and a ratio that remained at least at pretransplant levels throughout the study period. Seven participants (20%) developed anti-human leukocyte antigen antibodies without adversely affecting allograft function or histology on 2-year biopsies. Long-term follow-up is underway to assess the potential benefits of this regimen in children.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Antígenos HLA/imunologia , Transplante de Rim/imunologia , Linfócitos T/efeitos dos fármacos , Adolescente , Alemtuzumab , Criança , Feminino , Humanos , Terapia de Imunossupressão , Masculino , Estudos Prospectivos
13.
J Immunol ; 187(9): 4530-41, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21949023

RESUMO

Fetomaternal tolerance has been shown to depend both on regulatory T cells (Tregs) and negative signals from the PD1-PDL1 costimulatory pathway. More recently, IL-17-producing T cells (Th17) have been recognized as a barrier in inducing tolerance in transplantation. In this study, we investigate the mechanisms of PDL1-mediated regulation of fetomaternal tolerance using an alloantigen-specific CD4(+) TCR transgenic mouse model system (ABM-tg mouse). PDL1 blockade led to an increase in embryo resorption and a reduction in litter size. This was associated with a decrease in Tregs, leading to a lower Treg/effector T cell ratio. Moreover, PDL1 blockade inhibited Ag-specific alloreactive T cell apoptosis and induced apoptosis of Tregs and a shift toward higher frequency of Th17 cells, breaking fetomaternal tolerance. These Th17 cells arose predominantly from CD4(+)Foxp3(-) cells, rather than from conversion of Tregs. Locally in the placenta, similar decrease in regulatory and apoptotic markers was observed by real-time PCR. Neutralization of IL-17 abrogated the anti-PDL1 effect on fetal survival rate and restored Treg numbers. Finally, the adoptive transfer of Tregs was also able to improve fetal survival in the setting of PDL1 blockade. This is to our knowledge the first report using an alloantigen-specific model that establishes a link between PDL1, Th17 cells, and fetomaternal tolerance.


Assuntos
Antígeno B7-H1/fisiologia , Antígenos de Histocompatibilidade Classe II/fisiologia , Histocompatibilidade Materno-Fetal/imunologia , Tolerância Imunológica , Interleucina-17/fisiologia , Transdução de Sinais/imunologia , Células Th17/imunologia , Células Th17/metabolismo , Animais , Antígenos de Diferenciação/fisiologia , Antígeno B7-H1/antagonistas & inibidores , Feminino , Técnicas de Introdução de Genes , Antígenos de Histocompatibilidade Classe II/genética , Histocompatibilidade Materno-Fetal/genética , Tolerância Imunológica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Receptor de Morte Celular Programada 1/fisiologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/citologia , Células Th2/imunologia , Células Th2/metabolismo
14.
J Immunol ; 187(5): 2252-60, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21795594

RESUMO

Clinical trials using allogeneic mesenchymal stem cells (MSCs) are ongoing for the purpose of providing therapeutic benefit for a variety of human disorders. Pertinent to their clinical use are the accessibility to sufficient quantities of these cells allowing for repetitive administration, as well as a better understanding of the specific mechanisms by which allogeneic MSCs evade host immune responses that in turn influence their life span following administration. In this report, we sought to characterize and compare human peripheral blood MSCs (hPB-MSCs) with bone marrow-derived MSCs. hPB-MSCs met the established criteria to characterize this cellular lineage, including capacity for self-renewal, differentiation into tissues of mesodermal origin, and expression of phenotypic surface markers. In addition, hPB-MSCs suppressed alloreactive proliferation as well as the production of proinflammatory cytokines. Examination of the mechanisms by which allogeneic MSCs evade the host immune response, which is crucial for their therapeutic use, demonstrated that constitutive expression of serine protease inhibitor 9 (PI-9) on hPB-MSCs and bone marrow-derived MSCs is a major defense mechanism against granzyme B-mediated destruction by NK cells. Similarly, MSCs treated with small interfering RNA for PI-9 increased MSC cellular death, whereas expression of transgenic PI-9 following retroviral transduction protected MSCs. These data significantly advance our understanding of the immunomodulatory role for hPB-MSCs as well as the mechanisms by which they evade host immune responses. These findings contribute to the development of MSC-based therapies for diseases.


Assuntos
Citotoxicidade Imunológica/imunologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Serpinas/imunologia , Células da Medula Óssea/citologia , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/métodos , Diferenciação Celular/imunologia , Proliferação de Células , Separação Celular , Citometria de Fluxo , Humanos , Células Matadoras Naturais/imunologia , Transplante de Células-Tronco de Sangue Periférico/métodos , Serpinas/metabolismo
15.
J Immunol ; 187(3): 1097-105, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21697456

RESUMO

Programmed death-1 ligand 1 (PD-L1) is a coinhibitory molecule that negatively regulates multiple tolerance checkpoints. In the NOD mouse model, PD-L1 regulates the development of diabetes. PD-L1 has two binding partners, programmed death-1 and B7-1, but the significance of the PD-L1:B7-1 interaction in regulating self-reactive T cell responses is not yet clear. To investigate this issue in NOD mice, we have compared the effects of two anti-PD-L1 Abs that have different blocking activities. Anti-PD-L1 mAb 10F.2H11 sterically and functionally blocks only PD-L1:B7-1 interactions, whereas anti-PD-L1 mAb 10F.9G2 blocks both PD-L1:B7-1 and PD-L1:programmed death-1 interactions. Both Abs had potent, yet distinct effects in accelerating diabetes in NOD mice: the single-blocker 10F.2H11 mAb was more effective at precipitating diabetes in older (13-wk-old) than in younger (6- to 7-wk-old) mice, whereas the dual-blocker 10F.9G2 mAb rapidly induced diabetes in NOD mice of both ages. Similarly, 10F.2H11 accelerated diabetes in recipients of T cells from diabetic, but not prediabetic mice, whereas 10F.9G2 was effective in both settings. Both anti-PD-L1 mAbs precipitated diabetes in adoptive transfer models of CD4(+) and CD8(+) T cell-driven diabetes. Taken together, these data demonstrate that the PD-L1:B7-1 pathway inhibits potentially pathogenic self-reactive effector CD4(+) and CD8(+) T cell responses in vivo, and suggest that the immunoinhibitory functions of this pathway may be particularly important during the later phases of diabetogenesis.


Assuntos
Antígeno B7-1/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Inibidores do Crescimento/fisiologia , Glicoproteínas de Membrana/fisiologia , Peptídeos/fisiologia , Transdução de Sinais/imunologia , Transferência Adotiva , Animais , Anticorpos Bloqueadores/administração & dosagem , Antígenos CD/fisiologia , Proteínas Reguladoras de Apoptose/fisiologia , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Antígeno B7-H1 , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/transplante , Diabetes Mellitus Tipo 1/genética , Feminino , Ligantes , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Peptídeos/deficiência , Peptídeos/imunologia , Receptor de Morte Celular Programada 1 , Ligação Proteica/imunologia , Transdução de Sinais/genética
16.
Blood ; 117(4): 1176-83, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21076046

RESUMO

Clinical trials using mesenchymal stem cells (MSCs) have been initiated worldwide. An improved understanding of the mechanisms by which allogeneic MSCs evade host immune responses is paramount to regulating their survival after administration. This study has focused on the novel role of serine protease inhibitor (SPI) in the escape of MSCs from host immunosurveillance through the inhibition of granzyme B (GrB). Our data indicate bone marrow-derived murine MSCs express SPI6 constitutively. MSCs from mice deficient for SPI6 (SPI6(-/-)) exhibited a 4-fold higher death rate by primed allogeneic cytotoxic T cells than did wild-type MSCs. A GrB inhibitor rescued SPI6(-/-) MSCs from cytotoxic T-cell killing. Transduction of wild-type MSCs with MigR1-SPI6 also protected MSCs from cytotoxic T cell-mediated death in vitro. In addition, SPI6(-/-) MSCs displayed a shorter lifespan than wild-type MSCs when injected into an allogeneic host. We conclude that SPI6 protects MSCs from GrB-mediated killing and plays a pivotal role in their survival in vivo. Our data could serve as a basis for future SPI-based strategies to regulate the survival and function of MSCs after administration and to enhance the efficacy of MSC-based therapy for diseases.


Assuntos
Evasão da Resposta Imune/genética , Proteínas de Membrana/genética , Células-Tronco Mesenquimais/metabolismo , Serina Endopeptidases/genética , Serpinas/genética , Animais , Diferenciação Celular/genética , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Granzimas/antagonistas & inibidores , Granzimas/metabolismo , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Serina Endopeptidases/metabolismo , Inibidores de Serina Proteinase/genética , Inibidores de Serina Proteinase/metabolismo , Serpinas/metabolismo
17.
J Immunol ; 186(1): 121-31, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21131428

RESUMO

Antagonism of CXCR4 disrupts the interaction between the CXCR4 receptor on hematopoietic stem cells (HSCs) and the CXCL12 expressed by stromal cells in the bone marrow, which subsequently results in the shedding of HSCs to the periphery. Because of their profound immunomodulatory effects, HSCs have emerged as a promising therapeutic strategy for autoimmune disorders. We sought to investigate the immunomodulatory role of mobilized autologous HSCs, via target of the CXCR4-CXL12 axis, to promote engraftment of islet cell transplantation. Islets from BALB/c mice were transplanted beneath the kidney capsule of hyperglycemic C57BL/6 mice, and treatment of recipients with CXCR4 antagonist resulted in mobilization of HSCs and in prolongation of islet graft survival. Addition of rapamycin to anti-CXCR4 therapy further promoted HSC mobilization and islet allograft survival, inducing a robust and transferable host hyporesponsiveness, while administration of an ACK2 (anti-CD117) mAb halted CXCR4 antagonist-mediated HSC release and restored allograft rejection. Mobilized HSCs were shown to express high levels of the negative costimulatory molecule programmed death ligand 1 (PD-L1), and HSCs extracted from wild-type mice, but not from PD-L1 knockout mice, suppressed the in vitro alloimmune response. Moreover, HSC mobilization in PD-L1 knockout mice failed to prolong islet allograft survival. Targeting the CXCR4-CXCL12 axis thus mobilizes autologous HSCs and promotes long-term survival of islet allografts via a PD-L1-mediated mechanism.


Assuntos
Antígeno B7-1/fisiologia , Quimiocina CXCL12/antagonistas & inibidores , Marcação de Genes , Sobrevivência de Enxerto/imunologia , Transplante de Células-Tronco Hematopoéticas , Transplante das Ilhotas Pancreáticas/imunologia , Glicoproteínas de Membrana/fisiologia , Peptídeos/fisiologia , Receptores CXCR4/antagonistas & inibidores , Animais , Antígeno B7-1/genética , Antígeno B7-H1 , Benzilaminas , Quimiocina CXCL12/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Ciclamos , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/cirurgia , Marcação de Genes/métodos , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/genética , Compostos Heterocíclicos/farmacologia , Transplante das Ilhotas Pancreáticas/patologia , Masculino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Peptídeos/deficiência , Peptídeos/genética , Receptores CXCR4/metabolismo , Transplante Homólogo
18.
Diabetes ; 59(12): 3139-47, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20841611

RESUMO

OBJECTIVE: A number of clinical trials are underway to test whether mesenchymal stem cells (MSCs) are effective in treating various diseases, including type 1 diabetes. Although this cell therapy holds great promise, the optimal source of MSCs has yet to be determined with respect to major histocompatibility complex matching. Here, we examine this question by testing the ability of congenic MSCs, obtained from the NOR mouse strain, to reverse recent-onset type 1 diabetes in NOD mice, as well as determine the immunomodulatory effects of NOR MSCs in vivo. RESEARCH DESIGN AND METHODS: NOR MSCs were evaluated with regard to their in vitro immunomodulatory function in the context of autoreactive T-cell proliferation and dendritic cell (DC) generation. The in vivo effect of NOR MSC therapy on reversal of recent-onset hyperglycemia and on immunogenic cell subsets in NOD mice was also examined. RESULTS: NOR MSCs were shown to suppress diabetogenic T-cell proliferation via PD-L1 and to suppress generation of myeloid/inflammatory DCs predominantly through an IL-6-dependent mechanism. NOR MSC treatment of experimental type 1 diabetes resulted in long-term reversal of hyperglycemia, and therapy was shown to alter diabetogenic cytokine profile, to diminish T-cell effector frequency in the pancreatic lymph nodes, to alter antigen-presenting cell frequencies, and to augment the frequency of the plasmacytoid subset of DCs. CONCLUSIONS: These studies demonstrate the inimitable benefit of congenic MSC therapy in reversing experimental type 1 diabetes. These data should benefit future clinical trials using MSCs as treatment for type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Antígenos CD/análise , Antígenos CD/imunologia , Autoantígenos/imunologia , Diferenciação Celular , Citocinas/análise , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos NOD , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/patologia
19.
Clin Immunol ; 136(2): 174-87, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20452826

RESUMO

Induction of transplantation tolerance has the potential to allow for allograft acceptance without the need for life-long immunosuppression. Here we describe a novel approach that uses delivery of alloantigen by mature T cells to induce tolerance to fully allogeneic cardiac grafts. Adoptive transfer of mature alloantigen-expressing T cells into myeloablatively conditioned mice results in long-term acceptance of fully allogeneic heart transplants without evidence of chronic rejection. Since myeloablative conditioning is clinically undesirable we further demonstrated that adoptive transfer of mature alloantigen-expressing T cells alone into mice receiving non-myeloablative conditioning resulted in long-term acceptance of fully allogeneic heart allografts with minimal evidence of chronic rejection. Mechanistically, tolerance induction involved both deletion of donor-reactive host T cells and the development of regulatory T cells. Thus, delivery of alloantigen by mature T cells induces tolerance to fully allogeneic organ allografts in non-myeloablatively conditioned recipients, representing a novel approach for tolerance induction in transplantation.


Assuntos
Rejeição de Enxerto/prevenção & controle , Transplante de Coração/imunologia , Isoantígenos/administração & dosagem , Linfócitos T/imunologia , Tolerância ao Transplante/imunologia , Transplante Homólogo/imunologia , Transferência Adotiva , Animais , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Rejeição de Enxerto/imunologia , Isoantígenos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Fatores de Tempo , Irradiação Corporal Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA