Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34518219

RESUMEN

Triple-negative breast cancer (TNBC) is a breast cancer subtype that lacks targeted treatment options. The activation of the Notch developmental signaling pathway, which is a feature of TNBC, results in the secretion of proinflammatory cytokines and the recruitment of protumoral macrophages to the tumor microenvironment. While the Notch pathway is an obvious therapeutic target, its activity is ubiquitous, and predictably, anti-Notch therapies are burdened with significant on-target side effects. Previously, we discovered that, under conditions of cellular stress commonly found in the tumor microenvironment, the deubiquitinase USP9x forms a multiprotein complex with the pseudokinase tribbles homolog 3 (TRB3) that together activate the Notch pathway. Herein, we provide preclinical studies that support the potential of therapeutic USP9x inhibition to deactivate Notch. Using a murine TNBC model, we show that USP9x knockdown abrogates Notch activation, reducing the production of the proinflammatory cytokines, C-C motif chemokine ligand 2 (CCL2) and interleukin-1 beta (IL-1ß). Concomitant with these molecular changes, a reduction in tumor inflammation, the augmentation of antitumor immune response, and the suppression of tumor growth were observed. The pharmacological inhibition of USP9x using G9, a partially selective, small-molecule USP9x inhibitor, reduced Notch activity, remodeled the tumor immune landscape, and reduced tumor growth without associated toxicity. Proving the role of Notch, the ectopic expression of the activated Notch1 intracellular domain rescued G9-induced effects. This work supports the potential of USP9x inhibition to target Notch in metabolically vulnerable tissues like TNBC, while sparing normal Notch-dependent tissues.


Asunto(s)
Receptores Notch/genética , Transducción de Señal/genética , Neoplasias de la Mama Triple Negativas/genética , Ubiquitina Tiolesterasa/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Citocinas/genética , Regulación Neoplásica de la Expresión Génica/genética , Células HEK293 , Humanos , Interleucina-1beta/genética , Macrófagos/patología , Ratones , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral/genética
2.
J Immunol ; 203(11): 2791-2806, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31659015

RESUMEN

The paracaspase Malt1 is a key regulator of canonical NF-κB activation downstream of multiple receptors in both immune and nonimmune cells. Genetic disruption of Malt1 protease function in mice and MALT1 mutations in humans results in reduced regulatory T cells and a progressive multiorgan inflammatory pathology. In this study, we evaluated the altered immune homeostasis and autoimmune disease in Malt1 protease-deficient (Malt1PD) mice and the Ags driving disease manifestations. Our data indicate that B cell activation and IgG1/IgE production is triggered by microbial and dietary Ags preferentially in lymphoid organs draining mucosal barriers, likely as a result of dysregulated mucosal immune homeostasis. Conversely, the disease was driven by a polyclonal T cell population directed against self-antigens. Characterization of the Malt1PD T cell compartment revealed expansion of T effector memory cells and concomitant loss of a CD4+ T cell population that phenotypically resembles anergic T cells. Therefore, we propose that the compromised regulatory T cell compartment in Malt1PD animals prevents the efficient maintenance of anergy and supports the progressive expansion of pathogenic, IFN-γ-producing T cells. Overall, our data revealed a crucial role of the Malt1 protease for the maintenance of intestinal and systemic immune homeostasis, which might provide insights into the mechanisms underlying IPEX-related diseases associated with mutations in MALT1.


Asunto(s)
Autoinmunidad/inmunología , Homeostasis/inmunología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/inmunología , Linfocitos T Reguladores/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/deficiencia , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/genética
3.
Proc Natl Acad Sci U S A ; 108(4): 1555-60, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21205887

RESUMEN

14-3-3σ regulates cytokinesis and cell cycle arrest induced by DNA damage but its role in the immune system is unknown. Using gene-targeted 14-3-3σ-deficient (i.e., KO) mice, we studied the role of 14-3-3σ in B-cell functions. Total numbers of B cells were reduced by spontaneous apoptosis of peripheral B cells. Upon B-cell antigen receptor engagement in vitro, KO B cells did not proliferate properly or up-regulate CD86. In response to T cell-independent antigens, KO B cells showed poor secretion of antigen-specific IgM. This deficit led to increased lethality of KO mice after vesicular stomatitis virus infection. KO B cells showed elevated total FOXO transcriptional activity but also increased FOXO1 degradation. Coimmunoprecipitation revealed that endogenous 14-3-3σ protein formed a complex with FOXO1 protein. Our results suggest that 14-3-3σ maintains FOXO1 at a consistent level critical for normal B-cell antigen receptor signaling and B-cell survival.


Asunto(s)
Proteínas 14-3-3/inmunología , Linfocitos B/inmunología , Factores de Transcripción Forkhead/inmunología , Homeostasis/inmunología , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Traslado Adoptivo , Animales , Antígenos/inmunología , Apoptosis/inmunología , Linfocitos B/citología , Linfocitos B/metabolismo , Western Blotting , Proliferación Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Ficoll/análogos & derivados , Ficoll/inmunología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Receptores de Antígenos de Linfocitos B/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trinitrobencenos/inmunología
4.
Methods Cell Biol ; 188: 153-169, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38880522

RESUMEN

Pancreatic cancer remains an unmet medical need. Late diagnosis and the lack of efficient treatment significantly impact the prognosis of patients suffering from pancreatic cancer. Improving patient outcomes requires a deeper comprehension of the tumor ecosystem. To achieve this, a thorough exploration of the tumor microenvironment using pre-clinical models that accurately replicate human disease is imperative, particularly in understanding the dynamics of immune cell subsets. Surprisingly, the impact of model variations on the composition of the tumor microenvironment has been largely neglected. In this study, we introduce an orthotopic model of pancreatic ductal adenocarcinoma and a spontaneous model of insulinoma. Our findings reveal striking differences in the innate lymphoid cell infiltrate, highlighting the importance of considering model-specific influences when investigating the tumor microenvironment.


Asunto(s)
Carcinoma Ductal Pancreático , Modelos Animales de Enfermedad , Inmunidad Innata , Linfocitos , Neoplasias Pancreáticas , Microambiente Tumoral , Animales , Ratones , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/inmunología , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Microambiente Tumoral/inmunología , Linfocitos/inmunología , Humanos , Insulinoma/patología , Insulinoma/inmunología , Línea Celular Tumoral , Ratones Endogámicos C57BL
5.
Eur J Immunol ; 40(12): 3372-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21110320

RESUMEN

T-cell death is a fundamental process that is intricately regulated at multiple phases during T-cell differentiation, tolerance induction and the decline of the immune response. Caspase 3 is a crucial molecule regulating both mitochondrial and death receptor apoptotic pathways and therefore we were interested in examining the role of caspase 3 in T cells. Using P14 and H-Y CD8(+) TCR-transgenic models, our analysis has shown that caspase 3 is not required for thymic negative selection. In addition, caspase 3 does not play a prominent role in the contraction phase following acute viral infection, nor clonal deletion of CD8(+) T cells under tolerizing conditions. Surprisingly, our studies demonstrate that caspase 3 was not required for the induction of CD8(+) T-cell anergy in vivo, contrary to published reports using CD4(+) T cells. Therefore, these results demonstrate that caspase 3 is not essential in CD8(+) T cells for multiple forms of thymic or peripheral tolerance, nor the contraction phase after an acute anti-viral response.


Asunto(s)
Apoptosis , Linfocitos T CD8-positivos/metabolismo , Caspasa 3/metabolismo , Anergia Clonal , Timo/citología , Animales , Apoptosis/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Caspasa 3/inmunología , Diferenciación Celular/inmunología , Anergia Clonal/inmunología , Antígeno H-Y/inmunología , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/inmunología , Timo/crecimiento & desarrollo , Timo/inmunología
6.
Eur J Immunol ; 40(3): 867-77, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19950170

RESUMEN

Elucidating the signaling events that promote T-cell tolerance versus activation provides important insights for manipulating immunity in vivo. Previous studies have suggested that the absence of PKCtheta results in the induction of anergy and that the balance between the induction of the transcription factors NFAT, AP1 and NF-kappaB plays a key role in determining whether T-cell anergy or activation is induced. Here, we examine whether Bcl-10 and specific family members of NF-kappaB act downstream of PKCtheta to alter CD8(+) T-cell activation and/or anergy. We showed that T cells from mice deficient in c-Rel but not NF-kappaB1 (p50) have increased susceptibility to the induction of anergy, similar to T cells from PKCtheta-deficient mice. Surprisingly T cells from Bcl-10-deficient mice showed a strikingly different phenotype to the PKCtheta-deficient T cells, with a severe block in TCR-mediated activation. Furthermore, we have also shown that survival signals downstream of NF-kappaB, are uncoupled from signals that mediate T-cell anergy. These results suggest that c-Rel plays a critical role downstream of PKCtheta in controlling CD8(+) T-cell anergy induction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Linfocitos T CD8-positivos/inmunología , Anergia Clonal/inmunología , Isoenzimas/inmunología , Activación de Linfocitos/inmunología , Proteína Quinasa C/inmunología , Proteínas Proto-Oncogénicas c-rel/inmunología , Animales , Proteína 10 de la LLC-Linfoma de Células B , Western Blotting , Ratones , Ratones Transgénicos , FN-kappa B/inmunología , Fenotipo , Proteína Quinasa C-theta , Transducción de Señal/inmunología
7.
J Exp Med ; 195(4): 423-35, 2002 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-11854356

RESUMEN

Using a tumor model of spontaneously arising insulinomas expressing a defined tumor-associated antigen, we investigated whether tumor growth promotes cross-presentation and tolerance of tumor-specific T cells. We found that an advanced tumor burden enhanced cross-presentation of tumor-associated antigens to high avidity tumor-specific T cells, inducing T cell proliferation and limited effector function in vivo. However, contrary to other models, tumor-specific T cells were not tolerized despite a high tumor burden. In fact, in tumor-bearing mice, persistence and responsiveness of adoptively transferred tumor-specific T cells were enhanced. Accordingly, a potent T cell-mediated antitumor response could be elicited by intravenous administration of tumor-derived peptide and agonistic anti-CD40 antibody or viral immunization and reimmunization. Thus, in this model, tumor growth promotes activation of high avidity tumor-specific T cells instead of tolerance. Therefore, the host remains responsive to T cell immunotherapy.


Asunto(s)
Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Tolerancia Inmunológica , Insulinoma/inmunología , Insulinoma/patología , Activación de Linfocitos , Linfocitos T Citotóxicos/inmunología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/inmunología , Antígenos de Carbohidratos Asociados a Tumores/administración & dosificación , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Antígenos CD40/inmunología , División Celular , Citometría de Flujo , Receptores de Hialuranos/inmunología , Receptores de Hialuranos/metabolismo , Hipoglucemia/complicaciones , Vigilancia Inmunológica , Inmunoterapia Activa , Insulinoma/complicaciones , Insulinoma/terapia , Ganglios Linfáticos/inmunología , Ratones , Ratones Transgénicos , Quimera por Radiación , Análisis de Supervivencia , Linfocitos T Citotóxicos/citología , Factores de Tiempo
8.
Apoptosis ; 14(9): 1039-49, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19575295

RESUMEN

The p53-induced protein with a death domain, PIDD, was identified as a p53 target gene whose main role is to execute apoptosis in a p53-dependent manner. To investigate the physiological role of PIDD in apoptosis, we generated PIDD-deficient mice. Here, we report that, although PIDD expression is inducible upon DNA damage, PIDD-deficient mice undergo apoptosis normally not only in response to DNA damage, but also in response to various p53-independent stress signals and to death receptor (DR) engagement. This indicates that PIDD is not required for DNA damage-, stress-, and DR-induced apoptosis. Also, in the absence of PIDD, both caspase-2 processing and activation occur in response to DNA damage. Our findings demonstrate that PIDD does not play an essential role for all p53-mediated or p53-independent apoptotic pathways.


Asunto(s)
Apoptosis , Proteínas Portadoras/metabolismo , Daño del ADN , Estrés Fisiológico , Animales , Caspasa 2/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Marcación de Gen , Etiquetado Corte-Fin in Situ , Ratones , Procesamiento Proteico-Postraduccional , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Irradiación Corporal Total
9.
Mol Cell Biol ; 25(9): 3620-9, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15831467

RESUMEN

beta-Cell apoptosis is a key event contributing to the pathogenesis of type 1 diabetes mellitus. In addition to apoptosis being the main mechanism by which beta cells are destroyed, beta-cell apoptosis has been implicated in the initiation of type 1 diabetes mellitus through antigen cross-presentation mechanisms that lead to beta-cell-specific T-cell activation. Caspase-3 is the major effector caspase involved in apoptotic pathways. Despite evidence supporting the importance of beta-cell apoptosis in the pathogenesis of type 1 diabetes, the specific role of caspase-3 in this process is unknown. Here, we show that Caspase-3 knockout (Casp3(-/-) mice were protected from developing diabetes in a multiple-low-dose streptozotocin autoimmune diabetes model. Lymphocyte infiltration of the pancreatic islets was completely absent in Casp3(-/-) mice. To determine the role of caspase-3-dependent apoptosis in disease initiation, a defined antigen-T-cell receptor transgenic system, RIP-GP/P14 double-transgenic mice with Casp3 null mutation, was examined. beta-cell antigen-specific T-cell activation and proliferation were observed only in the pancreatic draining lymph node of RIP-GP/P14/Casp3(+/-) mice, but not in mice lacking caspase-3. Together, our findings demonstrate that caspase-3-mediated beta-cell apoptosis is a requisite step for T-cell priming, a key initiating event in type 1 diabetes.


Asunto(s)
Apoptosis , Caspasas/fisiología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Islotes Pancreáticos/patología , Animales , Apoptosis/genética , Caspasa 3 , Caspasas/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Glucagón/análisis , Glucagón/metabolismo , Insulina/análisis , Insulina/metabolismo , Islotes Pancreáticos/enzimología , Islotes Pancreáticos/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Pancrelipasa/inmunología , Linfocitos T/inmunología
10.
Cancer Discov ; 7(11): 1320-1335, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28790030

RESUMEN

Notch activation, which is associated with basal-like breast cancer (BLBC), normally directs tissue patterning, suggesting that it may shape the tumor microenvironment. Here, we show that Notch in tumor cells regulates the expression of two powerful proinflammatory cytokines, IL1ß and CCL2, and the recruitment of tumor-associated macrophages (TAM). Notch also regulates TGFß-mediated activation of tumor cells by TAMs, closing a Notch-dependent paracrine signaling loop between these two cell types. We use a mouse model in which Notch can be regulated in spontaneous mammary carcinoma to confirm that IL1ß and CCL2 production, and macrophage recruitment are Notch-dependent. In human disease, expression array analyses demonstrate a striking association between Notch activation, IL1ß and CCL2 production, macrophage infiltration, and BLBC. These findings place Notch at the nexus of a vicious cycle of macrophage infiltration and amplified cytokine secretion and provide immunotherapeutic opportunities in BLBC.Significance: BLBC is aggressive and has an unmet need for effective targeted treatment. Our data highlight immunotherapeutic opportunities in Notch-activated BLBC. Effective IL1ß and CCL2 antagonists are currently in clinical review to treat benign inflammatory disease, and their transition to the cancer clinic could have a rapid impact. Cancer Discov; 7(11); 1320-35. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1201.


Asunto(s)
Neoplasias de la Mama/genética , Quimiocina CCL2/genética , Interleucina-1beta/genética , Receptor Notch1/genética , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Quimiocina CCL2/inmunología , Quimiocina CCL2/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-1beta/inmunología , Interleucina-1beta/uso terapéutico , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Transgénicos , Comunicación Paracrina/genética , Receptor Notch1/inmunología , Receptor Notch1/uso terapéutico , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta/genética , Microambiente Tumoral/genética
11.
PLoS One ; 12(3): e0173176, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28257518

RESUMEN

Although the role of T cells in autoimmunity has been explored for many years, the mechanisms leading to the initial priming of an autoimmune T cell response remain enigmatic. The 'hit and run' model suggests that self-antigens released upon cell death can provide the initial signal for a self-sustaining autoimmune response. Using a novel transgenic mouse model where we could induce the release of self-antigens via caspase-dependent apoptosis. We tracked the fate of CD8+ T cells specific for the self-antigen. Our studies demonstrated that antigens released from apoptotic cells were cross-presented by CD11c+ cells in the draining lymph node. This cross-presentation led to proliferation of self-antigen specific T cells, followed by a transient ability to produce IFN-γ, but did not lead to the development of autoimmune diabetes. Using this model we examined the consequences on T cell immunity when apoptosis was combined with dendritic cell maturation signals, an autoimmune susceptible genetic background, and the deletion of Tregs. The results of our study demonstrate that autoimmune diabetes cannot be initiated by the presentation of antigens released from apoptotic cells in vivo even in the presence of factors known to promote autoimmunity.


Asunto(s)
Autoantígenos/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Linfocitos T Reguladores/inmunología , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Apoptosis/genética , Autoantígenos/inmunología , Autoinmunidad/genética , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Humanos , Tolerancia Inmunológica , Interferón gamma/genética , Ratones , Ratones Transgénicos/inmunología
12.
Cancer Cell ; 30(2): 337-348, 2016 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-27424808

RESUMEN

Mutations in the isocitrate dehydrogenase-1 gene (IDH1) are common drivers of acute myeloid leukemia (AML) but their mechanism is not fully understood. It is thought that IDH1 mutants act by inhibiting TET2 to alter DNA methylation, but there are significant unexplained clinical differences between IDH1- and TET2-mutant diseases. We have discovered that mice expressing endogenous mutant IDH1 have reduced numbers of hematopoietic stem cells (HSCs), in contrast to Tet2 knockout (TET2-KO) mice. Mutant IDH1 downregulates the DNA damage (DD) sensor ATM by altering histone methylation, leading to impaired DNA repair, increased sensitivity to DD, and reduced HSC self-renewal, independent of TET2. ATM expression is also decreased in human IDH1-mutated AML. These findings may have implications for treatment of IDH-mutant leukemia.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/enzimología , Isocitrato Deshidrogenasa/genética , Proteínas Proto-Oncogénicas/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Regulación hacia Abajo , Células Madre Hematopoyéticas/citología , Humanos , Isocitrato Deshidrogenasa/metabolismo , Ratones , Mutación , Proteínas Proto-Oncogénicas/metabolismo
13.
PLoS One ; 9(3): e92380, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24647761

RESUMEN

Vaccines for cancer immunotherapy are of interest but in general have not yet achieved the desired therapeutic efficacy in clinical trials. We present here a novel model to evaluate vaccine strategies by following tissue destruction in a transgenic model, where a defined antigen is expressed on pancreatic islets. We found that the transfer of syngeneic antigen-pulsed dendritic cells (DCs) resulted in autoimmune cytotoxic T-lymphocyte activation that was not observed following vaccinations that were based on peptides and adjuvants. Importantly, the induction of diabetes by DC transfer is dependent upon the maturation of DCs prior to transfer. Furthermore, diabetes induction only occurred if DCs were pulsed with the immunodominant epitope in addition to at least one other peptide, suggesting greater cytolytic activity upon engagement of multiple T-cell specificities. While the tumor environment undoubtedly will be more complex than healthy tissue, the insights gained through this model provide useful information on variables that can affect CD8-mediated tissue cytolysis in vivo.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Péptidos/farmacología , Animales , Células Cultivadas , Humanos , Ratones , Linfocitos T Citotóxicos/inmunología
14.
J Exp Med ; 210(7): 1419-31, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23797092

RESUMEN

The protein-tyrosine phosphatase Shp1 is expressed ubiquitously in hematopoietic cells and is generally viewed as a negative regulatory molecule. Mutations in Ptpn6, which encodes Shp1, result in widespread inflammation and premature death, known as the motheaten (me) phenotype. Previous studies identified Shp1 as a negative regulator of TCR signaling, but the severe systemic inflammation in me mice may have confounded our understanding of Shp1 function in T cell biology. To define the T cell­intrinsic role of Shp1, we characterized mice with a T cell­specific Shp1 deletion (Shp1fl/fl CD4-cre). Surprisingly, thymocyte selection and peripheral TCR sensitivity were unaltered in the absence of Shp1. Instead, Shp1(fl/fl) CD4-cre mice had increased frequencies of memory phenotype T cells that expressed elevated levels of CD44. Activation of Shp1-deficient CD4⁺ T cells also resulted in skewing to the Th2 lineage and increased IL-4 production. After IL-4 stimulation of Shp1- deficient T cells, Stat 6 activation was sustained, leading to enhanced Th2 skewing. Accordingly, we observed elevated serum IgE in the steady state. Blocking or genetic deletion of IL-4 in the absence of Shp1 resulted in a marked reduction of the CD44hi population. Therefore, Shp1 is an essential negative regulator of IL-4 signaling in T lymphocytes.


Asunto(s)
Homeostasis/inmunología , Interleucina-4/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Transducción de Señal/inmunología , Linfocitos T/enzimología , Linfocitos T/inmunología , Animales , Memoria Inmunológica , Interleucina-4/deficiencia , Interleucina-4/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Células Th2/enzimología , Células Th2/inmunología
15.
Nat Med ; 17(12): 1663-7, 2011 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-22081022

RESUMEN

Mature dendritic cells (DCs) are crucial for the induction of adaptive immune responses and perturbed DC homeostasis can result in autoimmune disease. Either uncontrolled expansion or enhanced survival of DCs can result in a variety of autoimmune diseases in mouse models. In addition, increased maturation signals, through overexpression of surface Toll-like receptors (TLRs) or stimulation by type I interferon (IFN), has been associated with systemic autoimmunity. Whereas recent studies have focused on identifying factors required for initiating the maturation process, the possibility that resting DCs also express molecules that 'hold' them in an immature state has generally not been considered. Here we show that nuclear factor-κB1 (NF-κB1) is crucial for maintaining the resting state of DCs. Self-antigen-pulsed unstimulated DCs that do not express NF-κB1 were able to activate CD8(+) T lymphocytes and induce autoimmunity. We further show that NF-κB1 negatively regulates the spontaneous production of tumor necrosis factor-α (TNF-α), which is associated with increased granzyme B expression in cytotoxic T lymphocytes (CTLs). These findings provide a new perspective on functional DC maturation and a potential mechanism that may account for pathologic T cell activation.


Asunto(s)
Inmunidad Adaptativa , Autoinmunidad , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , FN-kappa B/metabolismo , Animales , Autoantígenos/inmunología , Autoantígenos/metabolismo , Linfocitos T CD8-positivos/citología , Células Dendríticas/citología , Citometría de Flujo , Regulación de la Expresión Génica , Granzimas/metabolismo , Inmunohistoquímica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
16.
J Biol Chem ; 282(10): 7416-23, 2007 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-17213198

RESUMEN

In addition to its pro-apoptotic function in the death receptor pathway, roles for caspase-8 in mediating T-cell proliferation, maintaining lymphocyte homeostasis, and suppressing immunodeficiency have become evident. Humans with a germline point mutation of CASPASE-8 have multiple defects in T cells, B cells, and NK cells, most notably attenuated activation and immunodeficiency. By generating mice with B-cell-specific inactivation of caspase-8 (bcasp8(-/-)), we show that caspase-8 is dispensable for B-cell development, but its loss in B cells results in attenuated antibody production upon in vivo viral infection. We also report an important role for caspase-8 in maintaining B-cell survival following stimulation of the Toll-like receptor (TLR)2, -3, and -4. In response to TLR4 stimulation, caspase-8 is recruited to a complex containing IKKalphabeta, and its loss resulted in delayed NFkappaB nuclear translocation and impaired NFkappaB transcriptional activity. Our study supports dual roles for caspase-8 in apoptotic and nonapoptotic functions and demonstrates its requirement for TLR signaling and in the regulation of NFkappaB function.


Asunto(s)
Caspasa 8/fisiología , FN-kappa B/fisiología , Transducción de Señal/fisiología , Receptor Toll-Like 4/fisiología , Animales , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Infecciones por Rhabdoviridae/inmunología , Virus de la Estomatitis Vesicular Indiana , Receptor fas/fisiología
17.
Genes Dev ; 17(7): 883-95, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12654726

RESUMEN

Defects in death receptor-mediated apoptosis have been linked to cancer and autoimmune disease in humans. The in vivo role of caspase 8, a component of this pathway, has eluded analysis in postnatal tissues because of the lack of an appropriate animal model. Targeted disruption of caspase 8 is lethal in utero. We generated mice with a targeted caspase 8 mutation that is restricted to the T-cell lineage. Despite normal thymocyte development in the absence of caspase 8, we observed a marked decrease in the number of peripheral T-cells and impaired T-cell response ex vivo to activation stimuli. caspase 8 ablation protected thymocytes and activated T-cells from CD95 ligand but not anti-CD3-induced apoptosis, or apoptosis activated by agents that are known to act through the mitochondria. caspase 8 mutant mice were unable to mount an immune response to viral infection, indicating that caspase 8 deletion in T-cells leads to immunodeficiency. These findings identify an essential, cell-stage-specific role for caspase 8 in T-cell homeostasis and T-cell-mediated immunity. This is consistent with the recent identification of caspase 8 mutations in human immunodeficiency.


Asunto(s)
Caspasas/genética , Caspasas/metabolismo , Inmunidad Celular/fisiología , Activación de Linfocitos/genética , Linfocitos T/fisiología , Animales , Secuencia de Bases , Caspasa 8 , Caspasa 9 , Caspasas/deficiencia , Cartilla de ADN , Electroporación , Regulación del Desarrollo de la Expresión Génica , Genotipo , Homeostasis , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Linfocitos T/inmunología , Timidina Quinasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA