Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Autoimmunity ; 44(2): 137-48, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20695765

RESUMEN

The adipokine, leptin, regulates blood glucose and the insulin secretory function of beta cells, while also modulating immune cell function. We hypothesized that the dual effects of leptin may prevent or suppress the autoreactive destruction of beta cells in a virally induced rodent model of type 1 diabetes. Nearly 100% of weanling BBDR rats treated with the combination of an innate immune system activator, polyinosinic:polycytidylic acid (pIC), and Kilham rat virus (KRV) become diabetic within a predictable time frame. We utilized this model to test the efficacy of leptin in preventing diabetes onset, remitting new onset disease, and preventing autoimmune recurrence in diabetic rats transplanted with syngeneic islet grafts. High doses of leptin delivered via an adenovirus vector (AdLeptin) or alzet pump prevented diabetes in>90% of rats treated with pIC+KRV. The serum hyperleptinemia generated by this treatment was associated with decreased body weight, decreased non-fasting serum insulin levels, and lack of islet insulitis in leptin-treated rats. In new onset diabetics, hyperleptinemia prevented rapid weight loss and diabetic ketoacidosis, and temporarily restored euglycemia. Leptin treatment also prolonged the survival of syngeneic islets transplanted into diabetic BBDR rats. In diverse therapeutic settings, we found leptin treatment to have significant beneficial effects in modulating virally induced diabetes. These findings merit further evaluation of leptin as a potential adjunct therapeutic agent for treatment of human type 1 diabetes.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Leptina/uso terapéutico , Infecciones por Parvoviridae/inmunología , Parvovirus/inmunología , Animales , Glucemia , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/virología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/virología , Cetoacidosis Diabética/prevención & control , Humanos , Trasplante de Islotes Pancreáticos , Leptina/administración & dosificación , Leptina/inmunología , Infecciones por Parvoviridae/virología , Poli I-C/administración & dosificación , Poli I-C/inmunología , Ratas , Ratas Endogámicas BB , Resultado del Tratamiento
2.
PLoS One ; 5(7): e11812, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20676397

RESUMEN

The centrosome is important for microtubule organization and cell cycle progression in animal cells. Recently, mutations in the centrosomal protein, pericentrin, have been linked to human microcephalic osteodysplastic primordial dwarfism (MOPD II), a rare genetic disease characterized by severe growth retardation and early onset of type 2 diabetes among other clinical manifestations. While the link between centrosomal and cell cycle defects may account for growth deficiencies, the mechanism linking pericentrin mutations with dysregulated glucose homeostasis and pre-pubertal onset of diabetes is unknown. In this report we observed abundant expression of pericentrin in quiescent pancreatic beta-cells of normal animals which led us to hypothesize that pericentrin may have a critical function in beta-cells distinct from its known role in regulating cell cycle progression. In addition to the typical centrosome localization, pericentrin was also enriched with secretory vesicles in the cytoplasm. Pericentrin overexpression in beta-cells resulted in aggregation of insulin-containing secretory vesicles with cytoplasmic, but not centrosomal, pericentriolar material and an increase in total levels of intracellular insulin. RNAi- mediated silencing of pericentrin in secretory beta-cells caused dysregulated secretory vesicle hypersecretion of insulin into the media. Together, these data suggest that pericentrin may regulate the intracellular distribution and secretion of insulin. Mice transplanted with pericentrin-depleted islets exhibited abnormal fasting hypoglycemia and inability to regulate blood glucose normally during a glucose challenge, which is consistent with our in vitro data. This previously unrecognized function for a centrosomal protein to mediate vesicle docking in secretory endocrine cells emphasizes the adaptability of these scaffolding proteins to regulate diverse cellular processes and identifies a novel target for modulating regulated protein secretion in disorders such as diabetes.


Asunto(s)
Antígenos/metabolismo , Centrosoma/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Vesículas Secretoras/metabolismo , Animales , Antígenos/genética , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Células Secretoras de Insulina/ultraestructura , Masculino , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , ARN Interferente Pequeño/genética , Radioinmunoensayo , Vesículas Secretoras/ultraestructura
3.
Clin Immunol ; 135(1): 84-98, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20096637

RESUMEN

"Humanized" mouse models created by engraftment of immunodeficient mice with human hematolymphoid cells or tissues are an emerging technology with broad appeal across multiple biomedical disciplines. However, investigators wishing to utilize humanized mice with engrafted functional human immune systems are faced with a myriad of variables to consider. In this study, we analyze HSC engraftment methodologies using three immunodeficient mouse strains harboring the IL2rgamma(null) mutation; NOD-scid IL2rgamma(null), NOD-Rag1(null) IL2rgamma(null), and BALB/c-Rag1(null) IL2rgamma(null) mice. Strategies compared engraftment of human HSC derived from umbilical cord blood following intravenous injection into adult mice and intracardiac and intrahepatic injection into newborn mice. We observed that newborn recipients exhibited enhanced engraftment as compared to adult recipients. Irrespective of the protocol or age of recipient, both immunodeficient NOD strains support enhanced hematopoietic cell engraftment as compared to the BALB/c strain. Our data define key parameters for establishing humanized mouse models to study human immunity.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Subunidad gamma Común de Receptores de Interleucina/inmunología , Animales , Animales Recién Nacidos , Citometría de Flujo , Histocitoquímica , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Modelos Animales , Organismos Libres de Patógenos Específicos , Estadísticas no Paramétricas
4.
J Immunol ; 182(9): 5547-59, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19380803

RESUMEN

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.


Asunto(s)
Quimerismo , Trasplante de Células Madre Hematopoyéticas , Terapia de Inmunosupresión , Isoantígenos/genética , Lipopolisacáridos/administración & dosificación , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Receptores Toll-Like/agonistas , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Trasplante de Médula Ósea/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proteínas Portadoras/administración & dosificación , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Terapia de Inmunosupresión/métodos , Interferón Tipo I/biosíntesis , Interferón Tipo I/metabolismo , Interferón Tipo I/fisiología , Isoantígenos/inmunología , Isoantígenos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/fisiología , Poli I-C/administración & dosificación , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología , Trasplante de Piel/inmunología , Receptores Toll-Like/administración & dosificación , Receptores Toll-Like/metabolismo
5.
Diabetes ; 58(1): 165-73, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18984741

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Islotes Pancreáticos/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Ligando de CD40/inmunología , Citotoxicidad Inmunológica/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/cirugía , Citometría de Flujo , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/genética , Trasplante de Islotes Pancreáticos/métodos , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Ratones , Ratones Congénicos , Ratones Endogámicos C3H , Ratones Endogámicos NOD , Trasplante Homólogo
6.
Clin Dev Immunol ; 2008: 742810, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18815618

RESUMEN

Transplantation of allogeneic organs has proven to be an effective therapeutic for a large variety of disease states, but the chronic immunosuppression that is required for organ allograft survival increases the risk for infection and neoplasia and has direct organ toxicity. The establishment of transplantation tolerance, which obviates the need for chronic immunosuppression, is the ultimate goal in the field of transplantation. Many experimental approaches have been developed in animal models that permit long-term allograft survival in the absence of chronic immunosuppression. These approaches function by inducing peripheral or central tolerance to the allograft. Emerging as some of the most promising approaches for the induction of tolerance are protocols based on costimulation blockade. However, as these protocols move into the clinic, there is recognition that little is known as to their safety and efficacy when confronted with environmental perturbants such as virus infection. In animal models, it has been reported that virus infection can prevent the induction of tolerance by costimulation blockade and, in at least one experimental protocol, can lead to significant morbidity and mortality. In this review, we discuss how viruses modulate the induction and maintenance of transplantation tolerance.


Asunto(s)
Tolerancia al Trasplante/inmunología , Virosis/inmunología , Animales , Rechazo de Injerto/inmunología , Rechazo de Injerto/virología , Humanos , Inmunidad Innata/inmunología , Terapia de Inmunosupresión/métodos , Transducción de Señal/inmunología , Inmunología del Trasplante
7.
Exp Biol Med (Maywood) ; 233(8): 997-1012, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18653783

RESUMEN

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.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Animales , Antígenos CD34/sangre , Linfocitos B/citología , Linfocitos B/inmunología , Técnicas de Cultivo de Célula/métodos , Femenino , Hematopoyesis , Humanos , Recién Nacido , Subunidad gamma Común de Receptores de Interleucina/genética , Activación de Linfocitos , Linfopoyesis , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteínas Recombinantes/administración & dosificación , Linfocitos T/citología , Linfocitos T/inmunología , Acondicionamiento Pretrasplante , Trasplante Heterólogo , Factor de Necrosis Tumoral alfa/administración & dosificación
8.
Crit Rev Immunol ; 28(5): 403-39, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19166387

RESUMEN

Allogeneic organ transplantation has proven to be an effective therapeutic strategy for patients with end-stage organ disease. However, the chronic immunosuppression that is required for the survival of the allograft increases the risk for infection and malignancy. The establishment of transplantation tolerance, defined functionally as the survival of a donor allograft in the absence of immunosuppression, is the ultimate goal in the field of transplantation. Transplantation tolerance can be achieved using approaches that induce peripheral and/or central tolerance to the allograft. Protocols based on costimulation blockade (CB) have emerged as some of the most promising protocols for inducing long-term allograft survival in the absence of chronic immunosuppression. Despite its potential, recent evidence suggests that the efficacy of costimulation blockade can be reduced by environmental perturbations such as infection or inflammation, which activate Toll-like receptors (TLR). In this review, we discuss how the activation of TLRs can affect the induction and maintenance of transplantation tolerance.


Asunto(s)
Inmunidad Innata/inmunología , Tolerancia al Trasplante/inmunología , Animales , Rechazo de Injerto , Prueba de Histocompatibilidad , Humanos , Activación de Linfocitos , Receptores de Reconocimiento de Patrones/fisiología , Transducción de Señal , Linfocitos T/inmunología , Receptores Toll-Like/fisiología , Quimera por Trasplante , Trasplante Homólogo
9.
Clin Immunol ; 126(3): 303-14, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18096436

RESUMEN

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.


Asunto(s)
Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Islotes Pancreáticos/inmunología , Leucocitos Mononucleares/inmunología , Mutación/genética , Animales , Diabetes Mellitus/inmunología , Modelos Animales de Enfermedad , Rechazo de Injerto/inmunología , Humanos , Trasplante de Islotes Pancreáticos/inmunología , Leucocitos Mononucleares/trasplante , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Fenotipo
10.
Ann N Y Acad Sci ; 1150: 46-53, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19120266

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Modelos Animales de Enfermedad , Células Secretoras de Insulina/fisiología , Ratones , Animales , Autoinmunidad/fisiología , Diabetes Mellitus Tipo 1/inmunología , Humanos , Sistema Inmunológico/fisiología , Células Secretoras de Insulina/trasplante , Ratones SCID , Células Madre/fisiología , Trasplante Heterólogo/métodos
11.
J Immunol ; 178(2): 693-701, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17202329

RESUMEN

Viral infections are associated epidemiologically with the expression of type 1 diabetes in humans, but the mechanisms underlying this putative association are unknown. To investigate the role of viruses in diabetes, we used a model of viral induction of autoimmune diabetes in genetically susceptible biobreeding diabetes-resistant (BBDR) rats. BBDR rats do not develop diabetes in viral-Ab-free environments, but approximately 25% of animals infected with the parvovirus Kilham rat virus (KRV) develop autoimmune diabetes via a mechanism that does not involve beta cell infection. Using this model, we recently documented that TLR agonists synergize with KRV infection and increase disease penetrance. We now report that KRV itself activates innate immunity through TLR ligation. We show that KRV infection strongly stimulates BBDR splenocytes to produce the proinflammatory cytokines IL-6 and IL-12p40 but not TNF-alpha. KRV infection induces high levels of IL-12p40 by splenic B cells and Flt-3-ligand-induced bone marrow-derived dendritic cells (DCs) but only low levels of IL-12p40 production by thioglycolate-elicited peritoneal macrophages or GM-CSF plus IL-4-induced bone marrow-derived DCs. KRV-induced cytokine production is blocked by pharmacological inhibitors of protein kinase R and NF-kappaB. Genomic KRV DNA also induces BBDR splenocytes and Flt-3L-induced DCs from wild-type but not TLR9-deficient mice to produce IL-12p40; KRV-induced up-regulation of B lymphocytes can be blocked by TLR9 antagonists including inhibitory CpG and chloroquine. Administration of chloroquine to virus-infected BBDR rats decreases the incidence of diabetes and decreases blood levels of IL-12p40. Our data implicate the TLR9-signaling pathway in KRV-induced innate immune activation and autoimmune diabetes in the BBDR rat.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Salud , Parvovirus/inmunología , Transducción de Señal , Receptor Toll-Like 9/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Cruzamiento , Células Cultivadas , Cloroquina/farmacología , ADN Viral/genética , ADN Viral/aislamiento & purificación , Diabetes Mellitus Tipo 1/virología , Susceptibilidad a Enfermedades , Femenino , Genoma Viral/genética , Subunidad p40 de la Interleucina-12/biosíntesis , Subunidad p40 de la Interleucina-12/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Ratas , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Factor de Necrosis Tumoral alfa/metabolismo
12.
Blood ; 109(2): 819-26, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16973964

RESUMEN

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.


Asunto(s)
Rechazo de Injerto/inmunología , Linfocitos T/inmunología , Tolerancia al Trasplante , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Humanos , Interferón gamma/biosíntesis , Interferón gamma/sangre , Leucocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Trasplante de Piel/inmunología , Linfocitos T/citología , Factor de Necrosis Tumoral alfa/sangre
13.
Transplantation ; 82(8): 1085-92, 2006 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-17060858

RESUMEN

BACKGROUND: Blockade of T cell costimulation by treatment with donor-specific transfusion (DST) and anti-CD154 monoclonal antibody (mAb) induces prolonged allograft survival in mice. This effect is due in part to deletion of host CD8 and CD4 T cells that recognize alloantigen by direct presentation. The fate of host CD4 T cells that recognize alloantigen by indirect presentation, however, is unclear. METHODS: We studied Tg361 TCR transgenic CD4 T cells that recognize alloantigen by indirect presentation. Carboxyfluorescein diacetate, succinimidyl ester-labeled Tg361 cells were adoptively transferred into syngeneic nontransgenic recipients and their fate in the peripheral blood, spleen, and lymph nodes following treatment with DST and anti-CD154 was analyzed. RESULTS: Treatment of mice with DST plus anti-CD154 mAb does not delete Tg361 CD4 T cells, but instead renders them hyporesponsive to rechallenge with alloantigen. Mice circulating hyporesponsive CD4 T cells also fail to reject skin allografts. The hyporesponsive state of the T cells is not reversed by the addition of interleukin-2, anti-CD28 mAb, or an agonistic anti-CD134 mAb in the presence of antigen. These T cells are capable of activation, however, as evidenced by in vitro proliferation in response to anti-CD3 mAb. CONCLUSIONS: These results demonstrate that costimulation blockade can induce hyporesponsiveness of host CD4 T cells recognizing alloantigens by indirect presentation, thus prolonging graft survival by a mechanism that does not involve deletion of alloreactive T cells.


Asunto(s)
Presentación de Antígeno , Isoantígenos/química , Linfocitos T/citología , Animales , Linfocitos T CD4-Positivos/citología , Ligando de CD40/biosíntesis , Linfocitos T CD8-positivos/citología , Cricetinae , Fluoresceínas/farmacología , Supervivencia de Injerto , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Ratas
14.
Cell Metab ; 4(3): 245-54, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16950141

RESUMEN

In pancreatic beta cells, the endoplasmic reticulum (ER) is an important site for insulin biosynthesis and the folding of newly synthesized proinsulin. Here, we show that IRE1alpha, an ER-resident protein kinase, has a crucial function in insulin biosynthesis. IRE1alpha phosphorylation is coupled to insulin biosynthesis in response to transient exposure to high glucose; inactivation of IRE1alpha signaling by siRNA or inhibition of IRE1alpha phosphorylation hinders insulin biosynthesis. IRE1 activation by high glucose does not accompany XBP-1 splicing and BiP dissociation but upregulates its target genes such as WFS1. Thus, IRE1 signaling activated by transient exposure to high glucose uses a unique subset of downstream components and has a beneficial effect on pancreatic beta cells. In contrast, chronic exposure of beta cells to high glucose causes ER stress and hyperactivation of IRE1, leading to the suppression of insulin gene expression. IRE1 signaling is therefore a potential target for therapeutic regulation of insulin biosynthesis.


Asunto(s)
Retículo Endoplásmico/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/biosíntesis , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Chaperón BiP del Retículo Endoplásmico , Regulación de la Expresión Génica/fisiología , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Insulina/metabolismo , Secreción de Insulina , Proteínas de la Membrana/genética , Ratones , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Proinsulina/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Ratas , Factores de Transcripción del Factor Regulador X , Transducción de Señal/fisiología , Estrés Fisiológico/metabolismo , Estrés Fisiológico/fisiopatología , Factores de Transcripción , Regulación hacia Arriba/fisiología , Proteína 1 de Unión a la X-Box
15.
J Biol Chem ; 281(44): 33363-72, 2006 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-16931513

RESUMEN

NAD functions in multiple aspects of cellular metabolism and signaling through enzymes that covalently transfer ADP-ribose from NAD to acceptor proteins, thereby altering their function. NAD is a substrate for two enzyme families, mono-ADP-ribosyltransferases (mARTs) and poly(ADP-ribose) polymerases (PARPs), that covalently transfer an ADP-ribose monomer or polymer, respectively, to acceptor proteins. ART2, a mART, is a phenotypic marker of immunoregulatory cells found on the surface of T lymphocytes, including intestinal intraepithelial lymphocytes (IELs). We have shown that the auto-ADP-ribosylation of the ART2.2 allelic protein is multimeric. Our backbone structural alignment of ART2 (two alleles of the rat art2 gene have been reported, for simplicity, the ART2.2 protein investigated in this study will be referred to as ART2) and PARP suggested that multimeric auto-ADP-ribosylation of ART2 may represent an ADP-ribose polymer, rather than multiple sites of mono-ADP-ribosylation. To investigate this, we used highly purified recombinant ART2 and demonstrated that ART2 catalyzes the formation of an ADP-ribose polymer by sequencing gel and by HPLC and MS/MS mass spectrometry identification of PR-AMP, a breakdown product specific to poly(ADP-ribose). Furthermore, we identified the site of ADP-ribose polymer attachment on ART2 as Arg-185, an arginine in a crucial loop of its catalytic core. We found that endogenous ART2 on IELs undergoes multimeric auto-ADP-ribosylation more efficiently than ART2 on peripheral T cells, suggesting that these distinct lymphocyte populations differ in their ART2 surface topology. Furthermore, ART2.2 IELs are more resistant to NAD-induced cell death than ART2.1 IELs that do not have multimeric auto-ADP-ribosylation activity. The data suggest that capability of polymerizing ADP-ribose may not be unique to PARPs and that poly(ADP-ribosylation), an established nuclear activity, may occur extracellularly and modulate cell function.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , Membrana Celular/enzimología , Poli Adenosina Difosfato Ribosa/metabolismo , Linfocitos T/enzimología , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/genética , Adenosina Difosfato Ribosa/química , Adenosina Difosfato Ribosa/metabolismo , Animales , Catálisis , Supervivencia Celular , Modelos Moleculares , Hidrolasas Diéster Fosfóricas/metabolismo , Poli Adenosina Difosfato Ribosa/química , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Homología Estructural de Proteína
16.
Xenotransplantation ; 13(3): 224-32, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16756565

RESUMEN

BACKGROUND: The induction of xenogeneic hematopoietic chimerism is an attractive approach for overcoming the host response to xenografts, but establishing xenogeneic chimerism requires severe myeloablative conditioning of the recipient. The goal of this study was to determine if co-stimulation blockade would facilitate chimerism and xenograft tolerance in irradiation-conditioned concordant recipients. METHODS: Wistar Furth rat bone marrow (BM) cells were injected into irradiation-conditioned C57BL/6 mice with or without co-administration of anti-mouse CD154 monoclonal antibody (mAb). Chimerism was quantified by flow cytometry, and mice were transplanted with WF rat skin and islet xenografts. RESULTS: Blockade of CD40-CD154 interaction facilitated establishment of xenogeneic chimerism in mice conditioned with 600 cGy irradiation. Anti-CD154 mAb was not required for establishment of chimerism in mice treated with 700 cGy. However, mice irradiated with 700 cGy but not treated with anti-CD154 mAb developed a "graft-versus-host disease (GVHD)-like" wasting syndrome and died, irrespective of their development of chimerism. Xenogeneic chimeras established with irradiation and anti-CD154 mAb treatment exhibited prolonged skin and, in many cases, permanent islet xenograft survival. Chimerism was unstable and eventually lost in most recipients. Skin xenografts were rejected even in mice that remained chimeric, whereas most islet xenografts survived to the end of the observation period. CONCLUSIONS: Blockade of host CD40-CD154 interaction facilitates the establishment of xenogeneic chimerism and prevents wasting disease and death. Chimerism permits prolonged xenograft survival, but chimerism generated in this way is unstable over time. Skin xenografts are eventually rejected, whereas most islet xenografts survive long term and perhaps permanently.


Asunto(s)
Anticuerpos/farmacología , Trasplante de Médula Ósea/fisiología , Ligando de CD40/inmunología , Supervivencia de Injerto/fisiología , Hematopoyesis/inmunología , Trasplante de Islotes Pancreáticos/fisiología , Trasplante de Piel/fisiología , Quimera por Trasplante/inmunología , Trasplante Heterólogo/fisiología , Animales , Trasplante de Médula Ósea/patología , Antígenos CD40/inmunología , Citometría de Flujo , Reacción Huésped-Injerto , Trasplante de Islotes Pancreáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratas , Trasplante de Piel/patología , Ensayo de Capsula Subrrenal , Factores de Tiempo , Acondicionamiento Pretrasplante/métodos , Irradiación Corporal Total
17.
J Immunol ; 176(3): 1561-70, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16424185

RESUMEN

Costimulation blockade protocols are effective in prolonging allograft survival in animal models and are entering clinical trials, but how environmental perturbants affect graft survival remains largely unstudied. We used a costimulation blockade protocol consisting of a donor-specific transfusion and anti-CD154 mAb to address this question. We observed that lymphocytic choriomeningitis virus infection at the time of donor-specific transfusion and anti-CD154 mAb shortens allograft survival. Lymphocytic choriomeningitis virus 1) activates innate immunity, 2) induces allo-cross-reactive T cells, and 3) generates virus-specific responses, all of which may adversely affect allograft survival. To investigate the role of innate immunity, mice given costimulation blockade and skin allografts were coinjected with TLR2 (Pam3Cys), TLR3 (polyinosinic:polycytidylic acid), TLR4 (LPS), or TLR9 (CpG) agonists. Costimulation blockade prolonged skin allograft survival that was shortened after coinjection by TLR agonists. To investigate underlying mechanisms, we used "synchimeric" mice which circulate trace populations of anti-H2b transgenic alloreactive CD8+ T cells. In synchimeric mice treated with costimulation blockade, coadministration of all four TLR agonists prevented deletion of alloreactive CD8+ T cells and shortened skin allograft survival. These alloreactive CD8+ T cells 1) expressed the proliferation marker Ki-67, 2) up-regulated CD44, and 3) failed to undergo apoptosis. B6.TNFR2-/- and B6.IL-12R-/- mice treated with costimulation blockade plus LPS also exhibited short skin allograft survival whereas similarly treated B6.CD8alpha-/- and TLR4-/- mice exhibited prolonged allograft survival. We conclude that TLR signaling abrogates the effects of costimulation blockade by preventing alloreactive CD8+ T cell apoptosis through a mechanism not dependent on TNFR2 or IL-12R signaling.


Asunto(s)
Refuerzo Inmunológico de Injertos , Rechazo de Injerto/prevención & control , Inhibidores de Crecimiento/administración & dosificación , Trasplante de Piel/inmunología , Receptores Toll-Like/agonistas , Animales , Anticuerpos Monoclonales/administración & dosificación , Apoptosis/inmunología , Ligando de CD40/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Línea Celular Tumoral , Supresión Clonal/efectos de los fármacos , Femenino , Rechazo de Injerto/inmunología , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Quimera por Radiación , Receptores de Interleucina/metabolismo , Receptores de Interleucina-12 , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Receptor Toll-Like 4/metabolismo , Trasplante Homólogo
18.
Transplantation ; 78(11): 1601-8, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15591948

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Transfusión Sanguínea , Células de la Médula Ósea/fisiología , Ligando de CD40/fisiología , Supervivencia de Injerto , Trasplante de Piel , Animales , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/trasplante , Depleción Linfocítica , Metrizamida/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Bazo/citología , Trasplante Homólogo
19.
Transplantation ; 78(5): 660-7, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15371665

RESUMEN

BACKGROUND: Donor-specific transfusion (DST) and a brief course of anti-CD154 monoclonal antibody (mAb) induces permanent islet and prolonged skin allograft survival in mice. Induction of skin allograft survival requires the presence of CD4 cells and deletion of alloreactive CD8 cells. The specific roles of CD4 and CD4CD25 cells and the mechanism(s) by which they act are not fully understood. METHODS: We used skin and islet allografts, a CD8 T cell receptor (TCR) transgenic model system, and in vivo depleting antibodies to analyze the role of CD4 cell subsets in regulating allograft survival in mice treated with DST and anti-CD154 mAb. RESULTS: Deletion of CD4 or CD25 cells during costimulation blockade induced rapid rejection of skin but only minimally shortened islet allograft survival. Deletion of CD4 or CD25 cells had no effect upon survival of healed-in islet allografts, and CD25 cell deletion had no effect upon healed-in skin allograft survival. In the TCR transgenic model, DST plus anti-CD154 mAb treatment deleted alloreactive CD8 T cells, and anti-CD4 mAb treatment prevented that deletion. In contrast, injection of anti-CD25 mAb did not prevent alloreactive CD8 T cell deletion. CONCLUSIONS: These data document that (1) both CD4CD25 and CD4CD25 cells are required for induction of skin allograft survival, (2) CD4CD25 T cells are not required for alloreactive CD8 T cell deletion, and (3) CD4CD25 regulatory cells are not critical for islet allograft tolerance. It appears that skin and islet transplantation tolerance are mediated by different CD4 cell subsets and different mechanisms.


Asunto(s)
Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Piel/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Transfusión Sanguínea , Linfocitos T CD4-Positivos/inmunología , Ligando de CD40/inmunología , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Experimental/cirugía , Femenino , Terapia de Inmunosupresión/métodos , Activación de Linfocitos , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Modelos Animales , Trasplante Homólogo/inmunología
20.
Diabetes ; 53(8): 1972-8, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15277375

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Supervivencia de Injerto/fisiología , Trasplante de Islotes Pancreáticos/fisiología , Alelos , Animales , Ligando de CD40/inmunología , Terapia Combinada , Diabetes Mellitus Tipo 1/cirugía , Variación Genética , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA