Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
3.
PLoS One ; 7(11): e49213, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23155466

RESUMO

Despite increasing evidence that autoreactive CD8 T-cells are involved in both the initiation of type 1 diabetes (T1D) and the destruction of beta-cells, direct evidence for their destructive role in-vivo is lacking. To address a destructive role for autoreactive CD8 T-cells in human disease, we assessed the pathogenicity of a CD8 T-cell clone derived from a T1D donor and specific for an HLA-A2-restricted epitope of islet-specific glucose-6-phosphatase catalytic-subunit related protein (IGRP). HLA-A2/IGRP tetramer staining revealed a higher frequency of IGRP-specific CD8 T-cells in the peripheral blood of recent onset human individuals than of healthy donors. IGRP(265-273)-specific CD8 T-cells that were cloned from the peripheral blood of a recent onset T1D individual were shown to secrete IFNγ and Granzyme B after antigen-specific activation and lyse HLA-A2-expressing murine islets in-vitro. Lytic capacity was also demonstrated in-vivo by specific killing of peptide-pulsed target cells. Using the HLA-A2 NOD-scid IL2rγ(null) mouse model, HLA-A2-restricted IGRP-specific CD8 T-cells induced a destructive insulitis. Together, this is the first evidence that human HLA-restricted autoreactive CD8 T-cells target HLA-expressing beta-cells in-vivo, demonstrating the translational value of humanized mice to study mechanisms of disease and therapeutic intervention strategies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Antígeno HLA-A2/imunologia , Células Secretoras de Insulina/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Epitopos/imunologia , Epitopos/metabolismo , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Linfócitos T Citotóxicos/metabolismo
4.
PLoS One ; 4(10): e7251, 2009 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-19802382

RESUMO

BACKGROUND: The lack of a suitable animal model to study viral and immunological mechanisms of human dengue disease has been a deterrent to dengue research. METHODOLOGY/PRINCIPAL FINDINGS: We sought to establish an animal model for dengue virus (DENV) infection and immunity using non-obese diabetic/severe combined immunodeficiency interleukin-2 receptor gamma-chain knockout (NOD-scid IL2rgamma(null)) mice engrafted with human hematopoietic stem cells. Human CD45(+) cells in the bone marrow of engrafted mice were susceptible to in vitro infection using low passage clinical and established strains of DENV. Engrafted mice were infected with DENV type 2 by different routes and at multiple time points post infection, we detected DENV antigen and RNA in the sera, bone marrow, spleen and liver of infected engrafted mice. Anti-dengue IgM antibodies directed against the envelope protein of DENV peaked in the sera of mice at 1 week post infection. Human T cells that developed following engraftment of HLA-A2 transgenic NOD-scid IL2rgamma(null) mice with HLA-A2(+) human cord blood hematopoietic stem cells, were able to secrete IFN-gamma, IL-2 and TNF-alpha in response to stimulation with three previously identified A2 restricted dengue peptides NS4b 2353((111-119)), NS4b 2423((181-189)), and NS4a 2148((56-64)). CONCLUSIONS/SIGNIFICANCE: This is the first study to demonstrate infection of human cells and functional DENV-specific T cell responses in DENV-infected humanized mice. Overall, these mice should be a valuable tool to study the role of prior immunity on subsequent DENV infections.


Assuntos
Vírus da Dengue/imunologia , Sangue Fetal/virologia , Antígeno HLA-A2/imunologia , Hematopoese , Células-Tronco Hematopoéticas/virologia , Interleucina-2/imunologia , Camundongos SCID/imunologia , Animais , Dengue/imunologia , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Peptídeos/química , RNA/metabolismo
5.
J Immunol ; 182(9): 5547-59, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380803

RESUMO

Activation of TLR4 by administration of LPS shortens the survival of skin allografts in mice treated with costimulation blockade through a CD8 T cell-dependent, MyD88-dependent, and type I IFN receptor-dependent pathway. The effect of TLR activation on the establishment of allogeneic hematopoietic chimerism in mice treated with costimulation blockade is not known. Using a costimulation blockade protocol based on a donor-specific transfusion (DST) and a short course of anti-CD154 mAb, we show that LPS administration at the time of DST matures host alloantigen-presenting dendritic cells, prevents the establishment of mixed allogeneic hematopoietic chimerism, and shortens survival of donor-specific skin allografts. LPS mediates its effects via a mechanism that involves both CD4(+) and CD8(+) T cells and results from signaling through either the MyD88 or the type I IFN receptor pathways. We also document that timing of LPS administration is critical, as injection of LPS 24 h before treatment with DST and anti-CD154 mAb does not prevent hematopoietic engraftment but administration the day after bone marrow transplantation does. We conclude that TLR4 activation prevents the induction of mixed allogeneic hematopoietic chimerism through type I IFN receptor and MyD88-dependent signaling, which leads to the up-regulation of costimulatory molecules on host APCs and the generation of alloreactive T cells. These data suggest that distinct but overlapping cellular and molecular mechanisms control the ability of TLR agonists to block tolerance induction to hematopoietic and skin allografts in mice treated with costimulation blockade.


Assuntos
Quimerismo , Transplante de Células-Tronco Hematopoéticas , Terapia de Imunossupressão , Isoantígenos/genética , Lipopolissacarídeos/administração & dosagem , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Receptores Toll-Like/agonistas , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Transplante de Medula Óssea/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Proteínas de Transporte/administração & dosagem , Sobrevivência de Enxerto/genética , Sobrevivência de Enxerto/imunologia , Terapia de Imunossupressão/métodos , Interferon Tipo I/biossíntese , Interferon Tipo I/metabolismo , Interferon Tipo I/fisiologia , Isoantígenos/imunologia , Isoantígenos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/fisiologia , Poli I-C/administração & dosagem , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Transplante de Pele/imunologia , Receptores Toll-Like/administração & dosagem , Receptores Toll-Like/metabolismo
6.
J Immunol ; 182(5): 3278-84, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19234226

RESUMO

Fulminant inflammation in the liver is often accompanied by the accumulation of IFN-gamma-producing T cells. The BALB/c-Tgfb1(-/-) mouse exhibits extensive, spontaneously developing necroinflammation in the liver, accompanied by the accumulation of IFN-gamma-producing CD4(+) and CD8(+) T cells. Liver damage depends on the presence of an intact Ifng gene. We determined the relevant cellular source(s) of IFN-gamma. In Tgfb1(-/-) liver, CD4(+) T cells were more numerous than CD8(+) T cells and NK cells, and produced more IFN-gamma. Depletion of CD4(+) T cells eliminated both the elevation in plasma IFN-gamma and aspartate aminotransferase, whereas depletion of CD8(+) T cells did not. Rag1(-/-)Tgfb1(-/-) mice exhibited neither IFN-gamma elevation nor tissue damage, indicating that NK cells are not sufficient. IFN-gamma was required for strong overexpression of class II genes but not for CD4(+) T cell activation, oligoclonal expansion, or accumulation in the liver. The T cell inhibitory molecule PD-L1 was strongly expressed in Tgfb1(-/-) livers, ruling out a lack of PD-L1 expression as an explanation for aberrant liver T cell activation. Finally, whereas Tgfb1(-/-) CD4(+) T cells overexpressed Fas ligand, hepatocellular damage was observed in Fas(lpr/lpr)Tgfb1(-/-) mice, indicating that liver pathology is Fas independent. We conclude that liver damage in this model of fulminant autoimmune hepatitis is driven by CD4(+) T cell production of IFN-gamma, is independent of both CD8(+) T cells and the Fas ligand/Fas pathway, and is not explained by a lack of PD-L1 expression.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Hepatite Autoimune/imunologia , Hepatite Autoimune/patologia , Falência Hepática Aguda/imunologia , Falência Hepática Aguda/patologia , Receptor fas , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Modelos Animais de Doenças , Proteína Ligante Fas/genética , Proteína Ligante Fas/fisiologia , Hepatite Autoimune/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Interferon gama/deficiência , Interferon gama/genética , Interferon gama/fisiologia , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Falência Hepática Aguda/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Fator de Crescimento Transformador beta1/genética , Receptor fas/genética , Receptor fas/fisiologia
7.
Diabetes ; 58(1): 165-73, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18984741

RESUMO

OBJECTIVE: NOD mice model human type 1 diabetes and are used to investigate tolerance induction protocols for islet transplantation in a setting of autoimmunity. However, costimulation blockade-based tolerance protocols have failed in prolonging islet allograft survival in NOD mice. RESEARCH DESIGN AND METHODS: To investigate the underlying mechanisms, we studied the ability of costimulation blockade to prolong islet allograft survival in congenic NOD mice bearing insulin-dependent diabetes (Idd) loci that reduce the frequency of diabetes. RESULTS: The frequency of diabetes is reduced in NOD.B6 Idd3 mice and is virtually absent in NOD.B6/B10 Idd3 Idd5 mice. Islet allograft survival in NOD.B6 Idd3 mice treated with costimulation blockade is prolonged compared with NOD mice, and in NOD.B6/B10 Idd3 Idd5, mice islet allograft survival is similar to that achieved in C57BL/6 mice. Conversely, some Idd loci were not beneficial for the induction of transplantation tolerance. Alloreactive CD8 T-cell depletion in (NOD x CBA)F1 mice treated with costimulation blockade was impaired compared with similarly treated (C57BL/6.H2(g7) x CBA)F1 mice. Injection of exogenous interleukin (IL)-2 into NOD mice treated with costimulation prolonged islet allograft survival. NOD.B6 Idd3 mice treated with costimulation blockade deleted alloreactive CD8 T-cells and exhibited prolonged islet allograft survival. CONCLUSIONS: Il2 is the Idd3 diabetes susceptibility gene and can influence the outcome of T-cell deletion and islet allograft survival in mice treated with costimulation blockade. These data suggest that Idd loci can facilitate induction of transplantation tolerance by costimulation blockade and that IL-2/Idd3 is a critical component in this process.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Sobrevivência de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Ligante de CD40/imunologia , Citotoxicidade Imunológica/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/cirurgia , Citometria de Fluxo , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/genética , Transplante das Ilhotas Pancreáticas/métodos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos NOD , Transplante Homólogo
8.
Cell ; 134(4): 577-86, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18691745

RESUMO

Evaluation of the therapeutic potential of RNAi for HIV infection has been hampered by the challenges of siRNA delivery and lack of suitable animal models. Using a delivery method for T cells, we show that siRNA treatment can dramatically suppress HIV infection. A CD7-specific single-chain antibody was conjugated to oligo-9-arginine peptide (scFvCD7-9R) for T cell-specific siRNA delivery in NOD/SCIDIL2rgamma-/- mice reconstituted with human lymphocytes (Hu-PBL) or CD34+ hematopoietic stem cells (Hu-HSC). In HIV-infected Hu-PBL mice, treatment with anti-CCR5 (viral coreceptor) and antiviral siRNAs complexed to scFvCD7-9R controlled viral replication and prevented the disease-associated CD4 T cell loss. This treatment also suppressed endogenous virus and restored CD4 T cell counts in mice reconstituted with HIV+ peripheral blood mononuclear cells. Moreover, scFvCD7-9R could deliver antiviral siRNAs to naive T cells in Hu-HSC mice and effectively suppress viremia in infected mice. Thus, siRNA therapy for HIV infection appears to be feasible in a preclinical animal model.


Assuntos
Infecções por HIV/genética , Infecções por HIV/terapia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Linfócitos T/metabolismo , Animais , Antígenos CD7/metabolismo , Modelos Animais de Doenças , Expressão Gênica , HIV-1/genética , HIV-1/metabolismo , Humanos , Fragmentos de Imunoglobulinas/metabolismo , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , RNA Viral/metabolismo
9.
Exp Biol Med (Maywood) ; 233(8): 997-1012, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18653783

RESUMO

Umbilical cord blood (UCB) is increasingly being used for human hematopoietic stem cell (HSC) transplantation in children but often requires pooling multiple cords to obtain sufficient numbers for transplantation in adults. To overcome this limitation, we have used an ex vivo two-week culture system to expand the number of hematopoietic CD34(+) cells in cord blood. To assess the in vivo function of these expanded CD34(+) cells, cultured human UCB containing 1 x 10(6) CD34(+) cells were transplanted into conditioned NOD-scid IL2rgamma(null) mice. The expanded CD34(+) cells displayed short- and long-term repopulating cell activity. The cultured human cells differentiated into myeloid, B-lymphoid, and erythroid lineages, but not T lymphocytes. Administration of human recombinant TNFalpha to recipient mice immediately prior to transplantation promoted human thymocyte and T-cell development. These T cells proliferated vigorously in response to TCR cross-linking by anti-CD3 antibody. Engrafted TNFalpha-treated mice generated antibodies in response to T-dependent and T-independent immunization, which was enhanced when mice were co-treated with the B cell cytokine BLyS. Ex vivo expanded CD34(+) human UCB cells have the capacity to generate multiple hematopoietic lineages and a functional human immune system upon transplantation into TNFalpha-treated NOD-scid IL2rgamma(null) mice.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Subunidade gama Comum de Receptores de Interleucina/deficiência , Animais , Antígenos CD34/sangue , Linfócitos B/citologia , Linfócitos B/imunologia , Técnicas de Cultura de Células/métodos , Feminino , Hematopoese , Humanos , Recém-Nascido , Subunidade gama Comum de Receptores de Interleucina/genética , Ativação Linfocitária , Linfopoese , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Proteínas Recombinantes/administração & dosagem , Linfócitos T/citologia , Linfócitos T/imunologia , Condicionamento Pré-Transplante , Transplante Heterólogo , Fator de Necrose Tumoral alfa/administração & dosagem
10.
Curr Protoc Immunol ; Chapter 15: 15.21.1-15.21.21, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18491294

RESUMO

"Humanized" mice are a promising translational model for studying human hematopoiesis and immunity. Their utility has been enhanced by the development of new stocks of immunodeficient hosts, most notably mouse strains such as NOD-scid IL2rgamma(null) mice that lack the IL-2 receptor common gamma chain. These stocks of mice lack adaptive immune function, display multiple defects in innate immunity, and support heightened levels of human hematolymphoid engraftment. Humanized mice can support studies in many areas of immunology, including autoimmunity, transplantation, infectious diseases, and cancer. These models are particularly valuable in experimentation where there is no appropriate small animal model of the human disease, as in the case of certain viral infections. This unit details the creation of humanized mice by engraftment of immunodeficient mice with hematopoietic stem cells or peripheral blood mononuclear cells, provides methods for evaluating engraftment, and discusses considerations for choosing the appropriate model system to meet specific goals.


Assuntos
Hematopoese , Transplante de Células-Tronco Hematopoéticas , Imunidade Inata , Imunocompetência , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Modelos Animais , Transplante de Células-Tronco de Sangue Periférico , Animais , Animais Recém-Nascidos , Feminino , Citometria de Fluxo , Hematopoese/genética , Humanos , Imunidade Inata/genética , Imunocompetência/genética , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Baço/imunologia , Transplante Heterólogo
11.
Clin Immunol ; 126(3): 303-14, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18096436

RESUMO

Immunodeficient NOD-scid mice bearing a targeted mutation in the IL2 receptor common gamma chain (Il2rgamma(null)) readily engraft with human stem cells. Here we analyzed human peripheral blood mononuclear cells (PBMC) for their ability to engraft NOD-scid Il2rgamma(null) mice and established engraftment kinetics, optimal cell dose, and the influence of injection route. Even at low PBMC input, NOD-scid Il2rgamma(null) mice reproducibly support high human PBMC engraftment that plateaus within 3-4 weeks. In contrast to previous stocks of immunodeficient mice, we observed low intra- and inter-donor variability of engraftment. NOD-scid Il2rgamma(null) mice rendered hyperglycemic by streptozotocin treatment return to normoglycemia following transplantation with human islets. Interestingly, these human islet grafts are rejected following injection of HLA-mismatched human PBMC as evidenced by return to hyperglycemia and loss of human C-peptide. These data suggest that humanized NOD-scid Il2rgamma(null) mice may represent an important surrogate for investigating in vivo mechanisms of human islet allograft rejection.


Assuntos
Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Ilhotas Pancreáticas/imunologia , Leucócitos Mononucleares/imunologia , Mutação/genética , Animais , Diabetes Mellitus/imunologia , Modelos Animais de Doenças , Rejeição de Enxerto/imunologia , Humanos , Transplante das Ilhotas Pancreáticas/imunologia , Leucócitos Mononucleares/transplante , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Fenótipo
12.
Ann N Y Acad Sci ; 1150: 46-53, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19120266

RESUMO

Our understanding of the basic biology of diabetes has been guided by observations made using animal models, particularly rodents. However, humans are not mice, and outcomes predicted by murine studies are not always representative of actual outcomes in the clinic. In particular, investigators studying diabetes have relied heavily on mouse and rat models of autoimmune type 1-like diabetes, and experimental results using these models have not been representative of many of the clinical trials in type 1 diabetes. In this article, we describe the availability of new models of humanized mice for the study of three areas of diabetes. These include the use of humanized mice for the study of (1) human islet stem and progenitor cells, (2) human islet allograft rejection, and (3) human immunity and autoimmunity. These humanized mouse models provide an important preclinical bridge between in vitro studies and rodent models and the translation of discoveries in these model systems to the clinic.


Assuntos
Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Modelos Animais de Doenças , Células Secretoras de Insulina/fisiologia , Camundongos , Animais , Autoimunidade/fisiologia , Diabetes Mellitus Tipo 1/imunologia , Humanos , Sistema Imunitário/fisiologia , Células Secretoras de Insulina/transplante , Camundongos SCID , Células-Tronco/fisiologia , Transplante Heterólogo/métodos
13.
Ann N Y Acad Sci ; 1150: 149-51, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19120285

RESUMO

We investigated the mechanisms by which Toll-like receptor (TLR) agonists affect the induction of mixed chimerism and skin allograft survival in mice treated with co-stimulation blockade (CB). We report that TLR agonists prevent the generation of mixed chimerism by breaking tolerance in the alloreactive CD4(+) and CD8(+) T cell compartments, and that type I interferon (IFN) is important in this process. Understanding how environmental perturbations affect CB-induced transplantation tolerance may lead to more effective regimens that can be used as an approach for the treatment of type I diabetes, for which the transplantation of pancreatic islets is a promising therapy.


Assuntos
Quimerismo/efeitos dos fármacos , Imunossupressores/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Receptores Toll-Like/agonistas , Tolerância ao Transplante/efeitos dos fármacos , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Poli I-C/farmacologia , Receptor de Interferon alfa e beta/genética , Transplante de Pele/imunologia , Transplante de Pele/veterinária , Receptor 3 Toll-Like/agonistas , Receptor 4 Toll-Like/agonistas
14.
J Immunol ; 179(2): 786-96, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17617568

RESUMO

Delayed ICOS-B7h signal blockade promotes significant prolongation of cardiac allograft survival in wild-type but not in CD8-deficient C57BL/6 recipients of fully MHC-mismatched BALB/c heart allografts, suggesting the possible generation of CD8(+) regulatory T cells in vivo. We now show that the administration of a blocking anti-ICOS mAb results in the generation of regulatory CD8(+) T cells. These cells can transfer protection and prolong the survival of donor-specific BALB/c, but not third party C3H, heart grafts in CD8-deficient C57BL/6 recipients. This is unique to ICOS-B7h blockade, because B7 blockade by CTLA4-Ig prolongs graft survival in CD8-deficient mice and does not result in the generation of regulatory CD8(+) T cells. Those cells localize to the graft, produce both IFN-gamma and IL-4 after allostimulation in vitro, prohibit the expansion of alloreactive CD4(+) T cells, and appear to mediate a Th2 switch of recipient CD4(+) T cells after adoptive transfer in vivo. Finally, these cells are not confined to the CD28-negative population but express programmed death 1, a molecule required for their regulatory function in vivo. CD8(+)PD1(+) T cells suppress alloreactive CD4(+) T cells but do not inhibit the functions by alloreactive CD8(+) T cells in vitro. These results describe a novel allospecific regulatory CD8(+)PD1(+) T cell induced by ICOS-B7h blockade in vivo.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Linfócitos T CD8-Positivos/imunologia , Isoantígenos/imunologia , Proteínas/imunologia , Linfócitos T Reguladores/imunologia , Tolerância ao Transplante/imunologia , Transferência Adotiva , Animais , Citometria de Fluxo , Sobrevivência de Enxerto/imunologia , Transplante de Coração/imunologia , Ligante Coestimulador de Linfócitos T Induzíveis , Ativação Linfocitária/imunologia , Masculino , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/imunologia , Transplante Homólogo/imunologia
15.
Ann N Y Acad Sci ; 1103: 77-89, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17332083

RESUMO

There are many rodent models of autoimmune diabetes that have been used to study the pathogenesis of human type 1 diabetes (T1D), including the non-obese diabetic (NOD) mouse, the biobreeding (BB) rat, and the transgenic mouse models. However, mice and rats are not humans, and these rodent models do not completely recapitulate the autoimmune pathogenesis of the human disease. In addition, many of the reagents, tools, and therapeutics proposed for use in humans may be species specific and cannot be investigated in rodents. Researchers have used nonhuman primates to more closely mimic the human immune system and, to study species-specific therapeutics, but these studies are associated with additional ethical and economic constraints and, to date, no model of autoimmune diabetes in this species has been described. New animal models are needed that will permit the in vivo investigation of human immune systems and analyses of the pathogenesis of human T1D without putting individuals at risk. To fill this need, we are developing humanized mouse models for the in vivo study of T1D. These models are based on our newly generated stock of NOD-scid IL2rgamma(null) mice, which engraft at higher levels with human hematolymphoid cells and exhibit enhanced function of the engrafted human immune systems compared with previous humanized mouse models. Overall, development of these new generations of humanized mice should facilitate in vivo studies of the human immune system as well as permit the investigation of the pathogenesis and effector phases of human T1D.


Assuntos
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Animais , Modelos Animais de Doenças , Humanos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Reprodutibilidade dos Testes
16.
Ann N Y Acad Sci ; 1103: 90-3, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17376822

RESUMO

The use of "humanized" mice represents an appealing translational model for studies of the pathogenesis of immune-mediated diseases and for the evaluation of potential therapeutics. The utility of humanized mice depends on their ability to model the human immune system with high fidelity, and, in this respect, previous models have fallen short. The recently developed NOD-scid Il2rgamma(null) mouse, however, exhibits greatly enhanced ability to support the engraftment of human peripheral blood mononuclear cells. Herein, we describe the challenges of recapitulating human immunity in humanized mice and features of NOD-scid Il2rgamma(null) mice that help overcome them.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Ilhotas Pancreáticas/imunologia , Leucócitos Mononucleares/imunologia , Camundongos Endogâmicos NOD/genética , Camundongos SCID/genética , Animais , Cruzamentos Genéticos , Modelos Animais de Doenças , Humanos , Isoantígenos/imunologia , Camundongos , Camundongos Endogâmicos BALB C
17.
Blood ; 109(2): 819-26, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16973964

RESUMO

Allograft transplantation requires chronic immunosuppression, but there is no effective strategy to evaluate the long-term maintenance of immunosuppression other than assessment of graft function. The ability to monitor naive alloreactive T cells would provide an alternative guide for drug therapy at early, preclinical stages of graft rejection and for evaluating tolerance-inducing protocols. To detect and quantify naive alloreactive T cells directly ex vivo, we used the unique ability of naive T cells to rapidly produce TNF-alpha but not IFN-gamma. Naive alloreactive T cells were identified by the production of TNF-alpha after a 5-hour in vitro stimulation with alloantigen and were distinguished from effector/memory alloreactive T cells by the inability to produce IFN-gamma. Moreover, naive alloreactive T cells were not detected in mice tolerized against specific alloantigens. The frequency of TNF-alpha-producing cells was predictive for rejection in an in vivo cytotoxicity assay and correlated with skin allograft rejection. Naive alloreactive T cells were also detected in humans, suggesting clinical relevance. We conclude that rapid production of TNF-alpha can be used to quantify naive alloreactive T cells, that it is abrogated after the induction of tolerance, and that it is a potential tool to predict allograft rejection.


Assuntos
Rejeição de Enxerto/imunologia , Linfócitos T/imunologia , Tolerância ao Transplante , Fator de Necrose Tumoral alfa/biossíntese , Animais , Humanos , Interferon gama/biossíntese , Interferon gama/sangue , Leucócitos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Transplante de Pele/imunologia , Linfócitos T/citologia , Fator de Necrose Tumoral alfa/sangue
18.
J Immunol ; 177(10): 6675-84, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17082580

RESUMO

In both humans and NOD mice, particular combinations of MHC genes provide the primary risk factor for development of the autoreactive T cell responses causing type 1 diabetes (T1D). Conversely, other MHC variants can confer dominant T1D resistance, and previous studies in NOD mice have shown their expression on hemopoietically derived APC is sufficient to induce disease protection. Although allogeneic hemopoietic chimerization can clearly provide a means for blocking T1D development, its clinical use for this purpose has been obviated by a requirement to precondition the host with what would be a lethal irradiation dose if bone marrow engraftment is not successful. There have been reports in which T1D-protective allogeneic hemopoietic chimerization was established in NOD mice that were preconditioned by protocols not including a lethal dose of irradiation. In most of these studies, virtually all the hemopoietic cells in the NOD recipients eventually converted to donor type. We now report that a concern about such full allogeneic chimeras is that they are severely immunocompromised potentially because their T cells are positively selected in the thymus by MHC molecules differing from those expressed by the APC available in the periphery to activate T cell effector functions. However, this undesirable side effect of generalized immunosuppression is obviated by a new protocol that establishes without a lethal preconditioning component, a stable state of mixed allogeneic hemopoietic chimerism sufficient to inhibit T1D development and also induce donor-specific tolerance in NOD recipients.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Transplante de Células-Tronco Hematopoéticas , Quimera por Radiação/imunologia , Tolerância ao Transplante/imunologia , Animais , Linhagem Celular , Cricetinae , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/mortalidade , Feminino , Transplante de Células-Tronco Hematopoéticas/métodos , Hemocianinas/administração & dosagem , Hemocianinas/imunologia , Depleção Linfocítica , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Transplante de Pele/imunologia , Transplante de Pele/métodos , Timo/imunologia , Timo/patologia , Tolerância ao Transplante/genética
19.
Transplantation ; 78(11): 1601-8, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15591948

RESUMO

BACKGROUND: Treatment with anti-CD154 monoclonal antibody (mAb) plus a donor-specific transfusion (DST) of spleen cells prolongs skin allograft survival in mice through a mechanism involving deletion of host alloreactive CD8(+) T cells. It is unknown if other lymphohematopoietic cell populations can be used as a DST. METHODS: Murine recipients of allogeneic skin grafts on day 0 were either untreated or given a DST on day -7 plus 4 doses of anti-CD154 mAb on days -7, -4, 0, and +4. Deletion of CD8(+) alloreactive cells was measured using "synchimeric" CBA recipients, which circulate trace populations of TCR transgenic alloreactive CD8(+) T cells. RESULTS: Transfusion of splenocytes, thymocytes, lymph node cells, or buffy coat cells led to prolonged skin allograft survival in recipients treated with anti-CD154 mAb. In contrast, bone marrow DST failed to delete host alloreactive CD8(+) T cells and was associated with brief skin allograft survival. Transfusions consisting of bone marrow-derived dendritic cells or a mixture of splenocytes and bone marrow cells were also ineffective. CONCLUSIONS: Donor-specific transfusions of splenocytes, thymocytes, lymph node cells, or buffy coat cells can prolong skin allograft survival in recipients treated with costimulation blockade. Bone marrow cells fail to serve this function, in part by failing to delete host alloreactive CD8(+) T cells, and they may actively interfere with the function of a spleen cell DST. The data suggest that transplantation tolerance induction protocols that incorporate bone marrow cells to serve as a DST may not be effective.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Transfusão de Sangue , Células da Medula Óssea/fisiologia , Ligante de CD40/fisiologia , Sobrevivência de Enxerto , Transplante de Pele , Animais , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/transplante , Depleção Linfocítica , Metrizamida/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Baço/citologia , Transplante Homólogo
20.
Diabetes ; 53(8): 1972-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15277375

RESUMO

NOD mice develop type 1 autoimmune diabetes and exhibit genetically dominant resistance to transplantation tolerance induction. These two phenotypes are genetically separable. Costimulation blockade fails to prolong skin allograft survival in (NOD x C57BL/6)F1 mice and in NOD-related strains made diabetes-resistant by congenic introduction of protective major histocompatibility complex (MHC) or non-MHC Idd region genes. Here, we tested the hypothesis that the genetic basis for the resistance of NOD mice to skin allograft tolerance also applies to islet allografts. Surprisingly, costimulation blockade induced permanent islet allograft survival in (NOD x C57BL/6)F1 mice but not in NOD mice. After costimulation blockade, islet allograft survival was prolonged in diabetes-resistant NOD.B6 Idd3 mice and shortened in diabetes-free C57BL/6 mice congenic for the NOD Idd3 variant. Islet allograft tolerance could not be induced in diabetes-resistant NOD.B10 Idd5 and NOD.B10 Idd9 mice. The data demonstrate that 1) NOD mice resist islet allograft tolerance induction; 2) unlike skin allografts, resistance to islet allograft tolerance is a genetically recessive trait; 3) an Idd3 region gene(s) is an important determinant of islet allograft tolerance induction; and 4) there may be overlap in the mechanism by which the Idd3 resistance locus improves self-tolerance and the induction of allotolerance.


Assuntos
Diabetes Mellitus Tipo 1/genética , Sobrevivência de Enxerto/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Alelos , Animais , Ligante de CD40/imunologia , Terapia Combinada , Diabetes Mellitus Tipo 1/cirurgia , Variação Genética , Imunoterapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...