Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 45(4): 774-787, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27742544

RESUMEN

The transcription factor Foxo3 plays a crucial role in myeloid cell function but its role in lymphoid cells remains poorly defined. Here, we have shown that Foxo3 expression was increased after T cell receptor engagement and played a specific role in the polarization of CD4+ T cells toward pathogenic T helper 1 (Th1) cells producing interferon-γ (IFN-γ) and granulocyte monocyte colony stimulating factor (GM-CSF). Consequently, Foxo3-deficient mice exhibited reduced susceptibility to experimental autoimmune encephalomyelitis. At the molecular level, we identified Eomes as a direct target gene for Foxo3 in CD4+ T cells and we have shown that lentiviral-based overexpression of Eomes in Foxo3-deficient CD4+ T cells restored both IFN-γ and GM-CSF production. Thus, the Foxo3-Eomes pathway is central to achieve the complete specialized gene program required for pathogenic Th1 cell differentiation and development of neuroinflammation.


Asunto(s)
Diferenciación Celular/fisiología , Proteína Forkhead Box O3/metabolismo , Interleucina-1/metabolismo , Proteínas de Dominio T Box/metabolismo , Células TH1/metabolismo , Células TH1/patología , Factores de Transcripción/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Diferenciación Celular/inmunología , Línea Celular , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Proteína Forkhead Box O3/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células HEK293 , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-1/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas de Dominio T Box/inmunología , Células TH1/inmunología
3.
Immunity ; 42(2): 239-251, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25692700

RESUMEN

T follicular helper (Tfh) cells are essential in the induction of high-affinity, class-switched antibodies. The differentiation of Tfh cells is a multi-step process that depends upon the co-receptor ICOS and the activation of phosphoinositide-3 kinase leading to the expression of key Tfh cell genes. We report that ICOS signaling inactivates the transcription factor FOXO1, and a Foxo1 genetic deletion allowed for generation of Tfh cells with reduced dependence on ICOS ligand. Conversely, enforced nuclear localization of FOXO1 inhibited Tfh cell development even though ICOS was overexpressed. FOXO1 regulated Tfh cell differentiation through a broad program of gene expression exemplified by its negative regulation of Bcl6. Final differentiation to germinal center Tfh cells (GC-Tfh) was instead FOXO1 dependent as the Foxo1(-/-) GC-Tfh cell population was substantially reduced. We propose that ICOS signaling transiently inactivates FOXO1 to initiate a Tfh cell contingency that is completed in a FOXO1-dependent manner.


Asunto(s)
Diferenciación Celular/inmunología , Proteínas de Unión al ADN/biosíntesis , Factores de Transcripción Forkhead/genética , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Linfocitos T Colaboradores-Inductores/citología , Animales , Activación Enzimática , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6 , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología
4.
PLoS Biol ; 19(12): e3001478, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34941868

RESUMEN

Highly efficient gene conversion systems have the potential to facilitate the study of complex genetic traits using laboratory mice and, if implemented as a "gene drive," to limit loss of biodiversity and disease transmission caused by wild rodent populations. We previously showed that such a system of gene conversion from heterozygous to homozygous after a sequence targeted CRISPR/Cas9 double-strand DNA break (DSB) is feasible in the female mouse germline. In the male germline, however, all DSBs were instead repaired by end joining (EJ) mechanisms to form an "insertion/deletion" (indel) mutation. These observations suggested that timing Cas9 expression to coincide with meiosis I is critical to favor conditions when homologous chromosomes are aligned and interchromosomal homology-directed repair (HDR) mechanisms predominate. Here, using a Cas9 knock-in allele at the Spo11 locus, we show that meiotic expression of Cas9 does indeed mediate gene conversion in the male as well as in the female germline. However, the low frequency of both HDR and indel mutation in both male and female germlines suggests that Cas9 may be expressed from the Spo11 locus at levels too low for efficient DSB formation. We suggest that more robust Cas9 expression initiated during early meiosis I may improve the efficiency of gene conversion and further increase the rate of "super-mendelian" inheritance from both male and female mice.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Conversión Génica/genética , Edición Génica/métodos , Animales , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/genética , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Femenino , Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/genética , Ingeniería Genética/métodos , Células Germinativas/metabolismo , Masculino , Meiosis/genética , Ratones , ARN Guía de Kinetoplastida/genética , Reparación del ADN por Recombinación/genética
5.
J Immunol ; 209(6): 1118-1127, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35948398

RESUMEN

In response to an intracellular infectious agent, the immune system produces a specific cellular response as well as a T cell-dependent Ab response. Precursor T cells differentiate into effector T cells, including Th1 cells, and T follicular helper (TFH) cells. The latter cooperate with B cells to form germinal centers and induce the formation of Ab-forming plasmacytes. One major focal point for control of T cell differentiation is the transcription factor BCL6. In this study, we demonstrated that the Bcl6 gene is regulated by FOXO1-binding, cis-acting sequences located in a highly conserved region of the first Bcl6 intron. In both mouse and human T cells, deletion of the tandem FOXO1 binding sites increased the expression of BCL6 and enhanced the proportion of TFH cells. These results reveal a fundamental control point for cellular versus humoral immunity.


Asunto(s)
Proteínas Proto-Oncogénicas c-bcl-6 , Células T Auxiliares Foliculares , Linfocitos T Colaboradores-Inductores , Animales , Centro Germinal , Humanos , Intrones/genética , Ratones , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Factores de Transcripción/metabolismo
6.
Immunity ; 41(6): 934-46, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25526308

RESUMEN

Gradations in extracellular regulated kinase (ERK) signaling have been implicated in essentially every developmental checkpoint or differentiation process encountered by lymphocytes. Yet, despite intensive effort, the molecular basis by which differences in ERK activation specify alternative cell fates remains poorly understood. We report here that differential ERK signaling controls lymphoid-fate specification through an alternative mode of action. While ERK phosphorylates most substrates, such as RSK, by targeting them through its D-domain, this well-studied mode of ERK action was dispensable for development of γδ T cells. Instead, development of γδ T cells was dependent upon an alternative mode of action mediated by the DEF-binding pocket (DBP) of ERK. This domain enabled ERK to bind a distinct and select set of proteins required for specification of the γδ fate. These data provide the first in vivo demonstration for the role of DBP-mediated interactions in orchestrating alternate ERK-dependent developmental outcomes.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/inmunología , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Activación Enzimática/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Estabilidad Proteica , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Transducción de Señal/genética , Especificidad por Sustrato/genética
7.
Proc Natl Acad Sci U S A ; 117(41): 25667-25678, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-32978300

RESUMEN

Memory CD8 T cells provide durable protection against diverse intracellular pathogens and can be broadly segregated into distinct circulating and tissue-resident populations. Paradigmatic studies have demonstrated that circulating memory cells can be further divided into effector memory (Tem) and central memory (Tcm) populations based on discrete functional characteristics. Following resolution of infection, we identified a persisting antigen-specific CD8 T cell population that was terminally fated with potent effector function but maintained memory T cell qualities and conferred robust protection against reinfection. Notably, this terminally differentiated effector memory CD8 T cell population (terminal-Tem) was conflated within the conventional Tem population, prompting redefinition of the classical characteristics of Tem cells. Murine terminal-Tem were transcriptionally, functionally, and developmentally unique compared to Tem cells. Through mass cytometry and single-cell RNA sequencing (RNA-seq) analyses of human peripheral blood from healthy individuals, we also identified an analogous terminal-Tem population of CD8 T cells that was transcriptionally distinct from Tem and Tcm Key findings from this study show that parsing of terminal-Tem from conventionally defined Tem challenge the reported characteristics of Tem biology, including enhanced presence in lymphoid tissues, robust IL-2 production, and recall potential, greater than expected homeostatic fitness, refined transcription factor dependencies, and a distinct molecular phenotype. Classification of terminal-Tem and clarification of Tem biology hold broad implications for understanding the molecular regulation of memory cell states and harnessing immunological memory to improve immunotherapies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Memoria Inmunológica/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Linaje de la Célula/inmunología , Células Cultivadas , Humanos , Ratones
8.
Nat Immunol ; 10(10): 1057-63, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19701188

RESUMEN

A screen for increased longevity in Caenorhabditis elegans has identified a transcription factor that programs cells for resistance to oxidative stress, DNA repair and cell cycle control. The mammalian orthologs of this factor are referred to as 'Foxo' for 'Forkhead box', with the second 'o' in the name denoting a subfamily of four members related by sequence. This family of factors is regulated by growth factors, oxidative stress or nutrient deprivation. Thus, it might readily control the inflammatory conflagration associated with infection-driven lymphocyte proliferation. Surprisingly, the first insights into Foxo-mediated immune regulation have instead revealed direct control of highly specialized genes of the adaptive immune system.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/inmunología , Homeostasis/inmunología , Inmunidad , Animales , Factores de Transcripción Forkhead/genética , Humanos
9.
Nat Immunol ; 10(2): 176-84, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19136962

RESUMEN

Foxo transcription factors have a conserved role in the adaptation of cells and organisms to nutrient and growth factor availability. Here we show that Foxo1 has a crucial, nonredundant role in T cells. In naive T cells, Foxo1 controlled the expression of the adhesion molecule L-selectin, the chemokine receptor CCR7 and the transcription factor Klf2, and its deletion was sufficient to alter lymphocyte trafficking. Furthermore, Foxo1 deficiency resulted in a severe defect in interleukin 7 receptor alpha-chain (IL-7Ralpha) expression associated with its ability to bind an Il7r enhancer. Finally, growth factor withdrawal induced a Foxo1-dependent increase in Sell, Klf2 and Il7r expression. These data suggest that Foxo1 regulates the homeostasis and life span of naive T cells by sensing growth factor availability and regulating homing and survival signals.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Factores de Transcripción Forkhead/metabolismo , Selectina L/biosíntesis , Receptores CCR7/biosíntesis , Receptores de Interleucina-7/biosíntesis , Linfocitos T/metabolismo , Animales , Western Blotting , Diferenciación Celular/inmunología , Supervivencia Celular , Citometría de Flujo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Expresión Génica , Regulación de la Expresión Génica/inmunología , Homeostasis/inmunología , Inmunoprecipitación , Selectina L/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , ARN Mensajero/análisis , Receptores CCR7/inmunología , Receptores de Interleucina-7/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología , Linfocitos T/inmunología
10.
Nat Immunol ; 10(5): 504-13, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19363483

RESUMEN

Foxo transcription factors regulate cell cycle progression, cell survival and DNA-repair pathways. Here we demonstrate that deficiency in Foxo3 resulted in greater expansion of T cell populations after viral infection. This exaggerated expansion was not T cell intrinsic. Instead, it was caused by the enhanced capacity of Foxo3-deficient dendritic cells to sustain T cell viability by producing more interleukin 6. Stimulation of dendritic cells mediated by the coinhibitory molecule CTLA-4 induced nuclear localization of Foxo3, which in turn inhibited the production of interleukin 6 and tumor necrosis factor. Thus, Foxo3 acts to constrain the production of key inflammatory cytokines by dendritic cells and to control T cell survival.


Asunto(s)
Células Dendríticas/inmunología , Factores de Transcripción Forkhead/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos CD/inmunología , Antígenos CD/metabolismo , Infecciones por Arenaviridae/inmunología , Western Blotting , Antígeno CTLA-4 , Células Dendríticas/metabolismo , Citometría de Flujo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Congénicos , Ratones Transgénicos , Transporte de Proteínas/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
11.
Trends Immunol ; 38(12): 888-903, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28882454

RESUMEN

As we describe the immune system in ever more exquisite detail, we might find that no matter how successful, this approach will not be sufficient to understand the spread of infectious agents, their susceptibility to vaccine therapy, and human disease resistance. Compared with the strict reductionism practiced as a means of characterizing most biological processes, I propose that the progression and outcome of disease-causing host-parasite interactions will be more clearly understood through a focus on disease ecology.


Asunto(s)
Coinfección/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad , Infecciones/inmunología , Modelos Inmunológicos , Zoonosis/inmunología , Animales , Evolución Biológica , Transmisión de Enfermedad Infecciosa , Humanos , Virulencia , Factores de Virulencia
12.
J Immunol ; 200(4): 1335-1346, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29311359

RESUMEN

Caspase-8 (CASP8) is known as an executioner of apoptosis, but more recent studies have shown that it participates in the regulation of necroptosis and innate immunity. In this study, we show that CASP8 negatively regulates retinoic acid-inducible gene I (RIG-I) signaling such that, in its absence, stimulation of the RIG-I pathway in dendritic cells (DCs) produced modestly enhanced activation of IFN regulatory factor 3 with correspondingly greater amounts of proinflammatory cytokines. In addition, mice lacking DC-specific CASP8 (dcCasp8-/- mice) develop age-dependent symptoms of autoimmune disease characterized by hyperactive DCs and T cells, spleen and liver immunopathology, and the appearance of Th1-polarized CD4+ T cells. Such mice infected with chronic lymphocytic choriomeningitis virus, an RNA virus detected by RIG-I, mounted an enhanced lymphocytic choriomeningitis virus-specific immune response as measured by increased proportions of Ag-specific CD4+ T cells and multicytokine-producing CD4+ and CD8+ T cells. These results show that CASP8 subtly modulates DC maturation, which controls the spontaneous appearance of autoimmune T cells while simultaneously attenuating the acquired immune system and its potential to control a persistent viral infection.


Asunto(s)
Autoinmunidad/inmunología , Caspasa 8/inmunología , Células Dendríticas/inmunología , Virosis/inmunología , Animales , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/inmunología , Linfocitos T/inmunología
13.
Proc Natl Acad Sci U S A ; 114(42): E8865-E8874, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-28973925

RESUMEN

The factors and steps controlling postinfection CD8+ T cell terminal effector versus memory differentiation are incompletely understood. Whereas we found that naive TCF7 (alias "Tcf-1") expression is FOXO1 independent, early postinfection we report bimodal, FOXO1-dependent expression of the memory-essential transcription factor TCF7 in pathogen-specific CD8+ T cells. We determined the early postinfection TCF7high population is marked by low TIM3 expression and bears memory signature hallmarks before the appearance of established memory precursor marker CD127 (IL-7R). These cells exhibit diminished TBET, GZMB, mTOR signaling, and cell cycle progression. Day 5 postinfection, TCF7high cells express higher memory-associated BCL2 and EOMES, as well as increased accumulation potential and capacity to differentiate into memory phenotype cells. TCF7 retroviral transduction opposes GZMB expression and the formation of KLRG1pos phenotype cells, demonstrating an active role for TCF7 in extinguishing the effector program and forestalling terminal differentiation. Past the peak of the cellular immune response, we report a gradient of FOXO1 and TCF7 expression, which functions to oppose TBET and orchestrate a continuum of effector-to-memory phenotypes.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Proteína Forkhead Box O1/metabolismo , Memoria Inmunológica/fisiología , Animales , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Diferenciación Celular , Proteína Forkhead Box O1/genética , Granzimas/genética , Granzimas/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/genética , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/inmunología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Subunidad alfa del Receptor de Interleucina-7/inmunología , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Lectinas Tipo C , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Receptores Inmunológicos/metabolismo
14.
Immunity ; 33(6): 890-904, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21167754

RESUMEN

Foxo transcription factors integrate extrinsic signals to regulate cell division, differentiation and survival, and specific functions of lymphoid and myeloid cells. Here, we showed the absence of Foxo1 severely curtailed the development of Foxp3(+) regulatory T (Treg) cells and those that developed were nonfunctional in vivo. The loss of function included diminished CTLA-4 receptor expression as the Ctla4 gene was a direct target of Foxo1. T cell-specific loss of Foxo1 resulted in exocrine pancreatitis, hind limb paralysis, multiorgan lymphocyte infiltration, anti-nuclear antibodies and expanded germinal centers. Foxo-mediated control over Treg cell specification was further revealed by the inability of TGF-ß cytokine to suppress T-bet transcription factor in the absence of Foxo1, resulting in IFN-γ secretion. In addition, the absence of Foxo3 exacerbated the effects of the loss of Foxo1. Thus, Foxo transcription factors guide the contingencies of T cell differentiation and the specific functions of effector cell populations.


Asunto(s)
Antígenos CD/biosíntesis , Factores de Transcripción Forkhead/metabolismo , Proteínas de Dominio T Box/metabolismo , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Antígenos CD/genética , Autoinmunidad/genética , Antígeno CTLA-4 , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/inmunología , Tolerancia Inmunológica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Balance Th1 - Th2 , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
15.
Immunity ; 28(3): 297-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18342002

RESUMEN

In this issue of Immunity, He et al. (2008) establish the logic and circuitry that determine CD4-CD8 lineage specification. It all comes down to an eighty base pair silencer switch.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Timo/citología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Humanos , Timo/inmunología
16.
Nature ; 477(7364): 335-9, 2011 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-21921917

RESUMEN

Dysfunction of the intestinal epithelium is believed to result in the excessive translocation of commensal bacteria into the bowel wall that drives chronic mucosal inflammation in Crohn's disease, an incurable inflammatory bowel disease in humans characterized by inflammation of the terminal ileum. In healthy individuals, the intestinal epithelium maintains a physical barrier, established by the tight contact of cells. Moreover, specialized epithelial cells such as Paneth cells and goblet cells provide innate immune defence functions by secreting mucus and antimicrobial peptides, which hamper access and survival of bacteria adjacent to the epithelium. Epithelial cell death is a hallmark of intestinal inflammation and has been discussed as a possible pathogenic mechanism driving Crohn's disease in humans. However, the regulation of epithelial cell death and its role in intestinal homeostasis remain poorly understood. Here we demonstrate a critical role for caspase-8 in regulating necroptosis of intestinal epithelial cells (IECs) and terminal ileitis. Mice with a conditional deletion of caspase-8 in the intestinal epithelium (Casp8(ΔIEC)) spontaneously developed inflammatory lesions in the terminal ileum and were highly susceptible to colitis. Casp8(ΔIEC) mice lacked Paneth cells and showed reduced numbers of goblet cells, indicating dysregulated antimicrobial immune cell functions of the intestinal epithelium. Casp8(ΔIEC) mice showed increased cell death in the Paneth cell area of small intestinal crypts. Epithelial cell death was induced by tumour necrosis factor (TNF)-α, was associated with increased expression of receptor-interacting protein 3 (Rip3; also known as Ripk3) and could be inhibited on blockade of necroptosis. Lastly, we identified high levels of RIP3 in human Paneth cells and increased necroptosis in the terminal ileum of patients with Crohn's disease, suggesting a potential role of necroptosis in the pathogenesis of this disease. Together, our data demonstrate a critical function of caspase-8 in regulating intestinal homeostasis and in protecting IECs from TNF-α-induced necroptotic cell death.


Asunto(s)
Apoptosis , Caspasa 8/metabolismo , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Caspasa 8/genética , Colitis/enzimología , Colitis/inmunología , Colitis/metabolismo , Colitis/patología , Enfermedad de Crohn/enzimología , Enfermedad de Crohn/inmunología , Eliminación de Gen , Células Caliciformes/patología , Humanos , Técnicas In Vitro , Ratones , Necrosis , Células de Paneth/enzimología , Células de Paneth/inmunología , Células de Paneth/metabolismo , Células de Paneth/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
17.
Proc Natl Acad Sci U S A ; 111(46): 16466-71, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25362052

RESUMEN

Signaling from the T-cell receptor (TCR) conditions T-cell differentiation and activation, requiring exquisite sensitivity and discrimination. Using mass cytometry, a high-dimensional technique that can probe multiple signaling nodes at the single-cell level, we interrogate TCR signaling dynamics in control C57BL/6 and autoimmunity-prone nonobese diabetic (NOD) mice, which show ineffective ERK activation after TCR triggering. By quantitating signals at multiple steps along the signaling cascade and parsing the phosphorylation level of each node as a function of its predecessors, we show that a small impairment in initial pCD3ζ activation resonates farther down the signaling cascade and results in larger defects in activation of the ERK1/2-S6 and IκBα modules. This nonlinear property of TCR signaling networks, which magnifies small initial differences during signal propagation, also applies in cells from B6 mice activated at different levels of intensity. Impairment in pCD3ζ and pSLP76 is not a feedback consequence of a primary deficiency in ERK activation because no proximal signaling defect was observed in Erk2 KO T cells. These defects, which were manifest at all stages of T-cell differentiation from early thymic pre-T cells to memory T cells, may condition the imbalanced immunoregulation and tolerance in NOD T cells. More generally, this amplification of small initial differences in signal intensity may explain how T cells discriminate between closely related ligands and adopt strongly delineated cell fates.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/fisiología , Síndromes de Inmunodeficiencia/inmunología , Sistema de Señalización de MAP Quinasas/fisiología , Espectrometría de Masas/métodos , Receptores de Antígenos de Linfocitos T/fisiología , Análisis de la Célula Individual/métodos , Animales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Activación Enzimática , Variación Genética , Proteínas I-kappa B/metabolismo , Tolerancia Inmunológica , Inmunidad Celular , Memoria Inmunológica , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Linfopoyesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/deficiencia , Inhibidor NF-kappaB alfa , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores de Antígenos de Linfocitos T/análisis , Autotolerancia , Timo/citología , Timo/inmunología
18.
J Immunol ; 192(12): 5548-60, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24808358

RESUMEN

Caspase-8, an executioner enzyme in the death receptor pathway, was shown to initiate apoptosis and suppress necroptosis. In this study, we identify a novel, cell death-independent role for caspase-8 in dendritic cells (DCs): DC-specific expression of caspase-8 prevents the onset of systemic autoimmunity. Failure to express caspase-8 has no effect on the lifespan of DCs but instead leads to an enhanced intrinsic activation and, subsequently, more mature and autoreactive lymphocytes. Uncontrolled TLR activation in a RIPK1-dependent manner is responsible for the enhanced functionality of caspase-8-deficient DCs, because deletion of the TLR-signaling mediator, MyD88, ameliorates systemic autoimmunity induced by caspase-8 deficiency. Taken together, these data demonstrate that caspase-8 functions in a cell type-specific manner and acts uniquely in DCs to maintain tolerance.


Asunto(s)
Caspasa 8/inmunología , Células Dendríticas/inmunología , Tolerancia Inmunológica/fisiología , Factor 88 de Diferenciación Mieloide/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Transducción de Señal/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Caspasa 8/genética , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal/genética , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
19.
J Immunol ; 189(2): 721-31, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22675204

RESUMEN

Effective immune responses depend upon appropriate T cell differentiation in accord with the nature of an infectious agent, and the contingency of differentiation depends minimally on TCR, coreceptor, and cytokine signals. In this reverse genetic study, we show that the MAPK Erk2 is not essential for T cell proliferation in the presence of optimum costimulation. Instead, it has opposite effects on T-bet and Gata3 expression and, hence, on Th1 and Th2 differentiation. Alternatively, in the presence of TGF-ß, the Erk pathway suppresses a large program of gene expression, effectively limiting the differentiation of Foxp3(+) regulatory T cells. In the latter case, the mechanisms involved include suppression of Gata3 and Foxp3, induction of Tbx21, phosphorylation of Smad2,3, and possibly suppression of Socs2, a positive inducer of Stat5 signaling. Consequently, loss of Erk2 severely impeded Th1 differentiation while enhancing the development of Foxp3(+)-induced T regulatory cells. Selected profiles of gene expression under multiple conditions of T cell activation illustrate the opposing consequences of Erk pathway signaling.


Asunto(s)
Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/inmunología , Polaridad Celular/inmunología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Subgrupos de Linfocitos T/enzimología , Subgrupos de Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/virología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Polaridad Celular/genética , Proliferación Celular , Células Cultivadas , Factores de Transcripción Forkhead/biosíntesis , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa 1 Activada por Mitógenos/deficiencia , Proteína Quinasa 1 Activada por Mitógenos/genética , Subgrupos de Linfocitos T/virología , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/virología , Células TH1/enzimología , Células TH1/inmunología , Células TH1/virología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
20.
Immunol Rev ; 236: 41-53, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20636807

RESUMEN

Programmed cell death (PCD) occurs widely in species from every kingdom of life. It has been shown to be an integral aspect of development in multicellular organisms, and it is an essential component of the immune response to infectious agents. An analysis of the phylogenetic origin of PCD now shows that it evolved independently several times, and it is fundamental to basic cellular physiology. Undoubtedly, PCD pervades all life at every scale of analysis. These considerations provide a backdrop for understanding the complexity of intertwined, but independent, cell death programs that operate within the immune system. In particular, the contributions of apoptosis, autophagy, and necrosis in the resolution of an immune response are considered.


Asunto(s)
Apoptosis/inmunología , Autofagia/inmunología , Inmunidad/inmunología , Transducción de Señal/inmunología , Animales , Caspasa 8/metabolismo , Supervivencia Celular/inmunología , Humanos , Necrosis/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA