Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Immunity ; 38(4): 681-93, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23562159

RESUMEN

How innate lymphoid cells (ILCs) in the thymus and gut become specialized effectors is unclear. The prototypic innate-like γδ T cells (Tγδ17) are a major source of interleukin-17 (IL-17). We demonstrate that Tγδ17 cells are programmed by a gene regulatory network consisting of a quartet of high-mobility group (HMG) box transcription factors, SOX4, SOX13, TCF1, and LEF1, and not by conventional TCR signaling. SOX4 and SOX13 directly regulated the two requisite Tγδ17 cell-specific genes, Rorc and Blk, whereas TCF1 and LEF1 countered the SOX proteins and induced genes of alternate effector subsets. The T cell lineage specification factor TCF1 was also indispensable for the generation of IL-22 producing gut NKp46(+) ILCs and restrained cytokine production by lymphoid tissue inducer-like effectors. These results indicate that similar gene network architecture programs innate sources of IL-17, independent of anatomical origins.


Asunto(s)
Proteínas del Grupo de Alta Movilidad/metabolismo , Interleucina-17/biosíntesis , Intestinos/inmunología , Subgrupos Linfocitarios/inmunología , Linfocitos T/inmunología , Animales , Antígenos Ly/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Redes Reguladoras de Genes/inmunología , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Inmunidad Innata/genética , Interleucina-17/genética , Interleucinas/inmunología , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo , Transducción de Señal/inmunología , Activación Transcripcional/inmunología , Interleucina-22
2.
Dev Dyn ; 244(1): 43-55, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25329411

RESUMEN

BACKGROUND: Among the complexities of skeletal muscle differentiation is a temporal distinction in the onset of expression of different lineage-specific genes. The lineage-determining factor MyoD is bound to myogenic genes at the onset of differentiation whether gene activation is immediate or delayed. How temporal regulation of differentiation-specific genes is established remains unclear. RESULTS: Using embryonic tissue, we addressed the molecular differences in the organization of the myogenin and muscle creatine kinase (MCK) gene promoters by examining regulatory factor binding as a function of both time and spatial organization during somitogenesis. At the myogenin promoter, binding of the homeodomain factor Pbx1 coincided with H3 hyperacetylation and was followed by binding of co-activators that modulate chromatin structure. MyoD and myogenin binding occurred subsequently, demonstrating that Pbx1 facilitates chromatin remodeling and modification before myogenic regulatory factor binding. At the same time, the MCK promoter was bound by HDAC2 and MyoD, and activating histone marks were largely absent. The association of HDAC2 and MyoD was confirmed by co-immunoprecipitation, proximity ligation assay (PLA), and sequential ChIP. CONCLUSIONS: MyoD differentially promotes activated and repressed chromatin structures at myogenic genes early after the onset of skeletal muscle differentiation in the developing mouse embryo.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Desarrollo de Músculos/fisiología , Músculo Esquelético/embriología , Proteína MioD/metabolismo , Regiones Promotoras Genéticas/fisiología , Animales , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Histona Desacetilasa 2/biosíntesis , Histona Desacetilasa 2/genética , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Ratones , Músculo Esquelético/citología , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
3.
J Virol ; 88(17): 9490-503, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24942579

RESUMEN

UNLABELLED: Virus-specific CD8+ T cells in the lymphoid organs contract at the resolution of virus infections by apoptosis or by dissemination into peripheral tissues, and those residing in nonlymphoid organs, including the peritoneal cavity and fat pads, are more resistant to apoptosis than those in the spleen and lymph nodes. This stability of memory T cells in the nonlymphoid tissues may enhance protection to secondary challenges. Here, we show that lymphocytic choriomeningitis virus (LCMV)-specific CD8+ T cells in nonlymphoid tissues were enriched for memory precursors (expressing high levels of interleukin-7 receptor and low levels of killer cell lectin-like receptor G1 [IL-7Rhi KLRG1lo]) and had higher expression of CD27, CXCR3, and T cell factor-1 (TCF-1), each a marker that is individually correlated with decreased apoptosis. CD8+ T cells in the peritoneal cavity of TCF-1-deficient mice had decreased survival, suggesting a role for TCF-1 in promoting survival in the nonlymphoid tissues. CXCR3+ CD8+ T cells resisted apoptosis and accumulated in the lymph nodes of mice treated with FTY720, which blocks the export of lymph node cells into peripheral tissue. The peritoneal exudate cells (PEC) expressed increased amounts of CXCR3 ligands, CXCL9 and CXCL10, which may normally recruit these nonapoptotic cells from the lymph nodes. In addition, adoptive transfer of splenic CD8+ T cells into PEC or spleen environments showed that the peritoneal environment promoted survival of CD8+ T cells. Thus, intrinsic stability of T cells which are present in the nonlymphoid tissues along with preferential migration of apoptosis-resistant CD8+ T cells into peripheral sites and the availability of tissue-specific factors that enhance memory cell survival may collectively account for the tissue-dependent apoptotic differences. IMPORTANCE: Most infections are initiated at nonlymphoid tissue sites, and the presence of memory T cells in nonlymphoid tissues is critical for protective immunity in various viral infection models. Virus-specific CD8+ T cells in the nonlymphoid tissues are more resistant to apoptosis than those in lymphoid organs during the resolution and memory phase of the immune response to acute LCMV infection. Here, we investigated the mechanisms promoting stability of T cells in the nonlymphoid tissues. This increased resistance to apoptosis of virus-specific CD8+ T cells in nonlymphoid tissues was due to several factors. Nonlymphoid tissues were enriched in memory phenotype CD8+ T cells, which were intrinsically resistant to apoptosis irrespective of the tissue environment. Furthermore, apoptosis-resistant CD8+ T cells preferentially migrated into the nonlymphoid tissues, where the availability of tissue-specific factors may enhance memory cell survival. Our findings are relevant for the generation of long-lasting vaccines providing protection at peripheral infection sites.


Asunto(s)
Estructuras Animales/inmunología , Apoptosis , Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/fisiología , Virus de la Coriomeningitis Linfocítica/inmunología , Subgrupos de Linfocitos T/fisiología , Estructuras Animales/patología , Animales , Antígenos de Superficie/análisis , Infecciones por Arenaviridae/patología , Linfocitos T CD8-positivos/química , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular , Masculino , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/química , Subgrupos de Linfocitos T/inmunología
4.
J Immunol ; 190(6): 2659-69, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23378428

RESUMEN

The Tec family tyrosine kinase, Itk, regulates signaling downstream of the TCR. The absence of Itk in CD4(+) T cells results in impaired Th2 responses along with defects in maturation, cytokine production, and survival of iNKT cells. Paradoxically, Itk(-/-) mice have spontaneously elevated serum IgE levels, resulting from an expansion of the Vγ1.1(+)Vδ6.3(+) subset of γδ T cells, known as γδ NKT cells. Comparisons between γδ NKT cells and αß iNKT cells showed convergence in the pattern of cell surface marker expression, cytokine profiles, and gene expression, suggesting that these two subsets of NKT cells undergo similar differentiation programs. Hepatic γδ NKT cells have an invariant TCR and are derived predominantly from fetal progenitors that expand in the thymus during the first weeks of life. The adult thymus contains these invariant γδ NKT cells plus a heterogeneous population of Vγ1.1(+)Vδ6.3(+) T cells with diverse CDR3 sequences. This latter population, normally excluded from the liver, escapes the thymus and homes to the liver when Itk is absent. In addition, Itk(-/-) γδ NKT cells persistently express high levels of Zbtb16 (PLZF) and Il4, genes that are normally downregulated in the most mature subsets of NKT cells. These data indicate that Itk signaling is required to prevent the expansion of γδ NKT cells in the adult thymus, to block their emigration, and to promote terminal NKT cell maturation.


Asunto(s)
Diferenciación Celular/inmunología , Senescencia Celular/inmunología , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Proteínas Tirosina Quinasas/fisiología , Receptores de Antígenos de Linfocitos T gamma-delta/biosíntesis , Timo/enzimología , Timo/inmunología , Animales , Inhibición de Migración Celular/inmunología , Movimiento Celular/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/citología , Timo/citología
5.
J Immunol ; 182(6): 3380-9, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265115

RESUMEN

The maturation of naive CD8(+) T cells into effector CTLs is a critical feature of a functional adaptive immune system. Development of CTLs depends, in part, upon the expression of the transcriptional regulator eomesodermin (EOMES), which is thought to regulate expression of two key effector molecules, perforin and granzyme B. Although EOMES is important for effector CTL development, the precise mechanisms regulating CD8(+) effector cell maturation remains poorly understood. In this study, we show that Notch1 regulates the expression of EOMES, perforin, and granzyme B through direct binding to the promoters of these crucial effector molecules. By abrogating Notch signaling, both biochemically as well as genetically, we conclude that Notch activity mediates CTL activity through direct regulation of EOMES, perforin, and granzyme B.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citotoxicidad Inmunológica , Receptor Notch1/fisiología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/fisiología , Animales , Diferenciación Celular/inmunología , Granzimas/antagonistas & inhibidores , Granzimas/genética , Granzimas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Perforina/antagonistas & inhibidores , Perforina/genética , Perforina/metabolismo , Regiones Promotoras Genéticas/inmunología , Unión Proteica/inmunología , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteínas de Dominio T Box/antagonistas & inhibidores , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
6.
Immunohorizons ; 3(7): 341-351, 2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31356163

RESUMEN

γδNKT cells are an abundant γδT cell population with restricted Vγ1.1 Vδ6.3 gene usage and phenotypic and functional similarity to conventional αß-invariant NKT cells. The γδNKT population responds to Listeria infections, but specific ligands are not known. In this work, we studied the CDR3 requirements of the γδNKT TCR, Vγ1.1Vδ6.3 for recognizing naive macrophages, and macrophages infected with Listeria We expressed four different variants of the Vγ1.1Vδ6.3 TCR in TCR-deficient hybridomas, one with germline-encoded sequences and three with nongermline-encoded sequences. All of the hybridomas were activated when cultured in the presence of macrophages, and the activation was increased when the macrophages were infected with Listeria This indicates that these TCRs can recognize a self-ligand present in macrophages and suggests that the ligand is modified or upregulated when the cells are infected with Listeria One of the three nongermline-encoded Vγ1.1 variants induced a lower activation level compared with the other variants tested in this study, suggesting that recognition of the Listeria-induced ligand involves the CDR3γ region of the TCR.


Asunto(s)
Regiones Determinantes de Complementariedad/genética , Células Germinativas/química , Listeria/inmunología , Listeriosis/microbiología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Genes Codificadores de la Cadena delta de los Receptores de Linfocito T/genética , Genes Codificadores de la Cadena gamma de los Receptores de Linfocito T/genética , Hibridomas/inmunología , Hibridomas/microbiología , Interleucina-2/metabolismo , Linfocitos Intraepiteliales/inmunología , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Células T Asesinas Naturales/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/clasificación , Transfección
7.
Front Immunol ; 5: 249, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24904593

RESUMEN

T cell stimulation requires the input and integration of external signals. Signaling through the T cell receptor (TCR) is known to induce formation of the membrane-tethered CBM complex, comprising CARMA1, BCL10, and MALT1, which is required for TCR-mediated NF-κB activation. TCR signaling has been shown to activate NOTCH proteins, transmembrane receptors also implicated in NF-κB activation. However, the link between TCR-mediated NOTCH signaling and early events leading to induction of NF-κB activity remains unclear. In this report, we demonstrate a novel cytosolic function for NOTCH1 and show that it is essential to CBM complex formation. Using a model of skin allograft rejection, we show in vivo that NOTCH1 acts in the same functional pathway as PKCθ, a T cell-specific kinase important for CBM assembly and classical NF-κB activation. We further demonstrate in vitro NOTCH1 associates physically with PKCθ and CARMA1 in the cytosol. Unexpectedly, when NOTCH1 expression was abrogated using RNAi approaches, interactions between CARMA1, BCL10, and MALT1 were lost. This failure in CBM assembly reduced inhibitor of kappa B alpha phosphorylation and diminished NF-κB-DNA binding. Finally, using a luciferase gene reporter assay, we show the intracellular domain of NOTCH1 can initiate robust NF-κB activity in stimulated T cells, even when NOTCH1 is excluded from the nucleus through modifications that restrict it to the cytoplasm or hold it tethered to the membrane. Collectively, these observations provide evidence that NOTCH1 may facilitate early events during T cell activation by nucleating the CBM complex and initiating NF-κB signaling.

8.
J Vis Exp ; (50)2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559006

RESUMEN

Chromatin immunoprecipitation (ChIP) is a powerful tool to identify protein:chromatin interactions that occur in the context of living cells. This technique has been widely exploited in tissue culture cells, and to a lesser extent, in primary tissue. The application of ChIP to rodent embryonic tissue, especially at early times of development, is complicated by the limited amount of tissue and the heterogeneity of cell and tissue types in the embryo. Here we present a method to perform ChIP using a dissociated embryonic day 8.5 (E8.5) embryo. Sheared chromatin from a single E8.5 embryo can be divided into up to five aliquots, which allows the investigator sufficient material for controls and for investigation of specific protein:chromatin interactions. We have utilized this technique to begin to document protein:chromatin interactions during the specification of tissue-specific gene expression programs. The heterogeneity of cell types in an embryo necessarily restricts the application of this technique because the result is the detection of protein:chromatin interactions without distinguishing whether the interactions occur in all, a subset of, or a single cell type(s). However, examination of tissue-specific genes during or following the onset of tissue-specific gene expression is feasible for two reasons. First, immunoprecipitation of tissue specific factors necessarily isolates chromatin from the cell type where the factor is expressed. Second, immunoprecipitation of coactivators and histones containing post-translational modifications that are associated with gene activation should only be found at genes and gene regulatory sequences in the cell type where the gene is being or has been activated. The technique should be applicable to the study of most tissue-specific gene activation events. In the example described below, we utilized E8.5 and E9.5 mouse embryos to examine factor binding at a skeletal muscle specific gene promoter. Somites, which are the precursor tissues from which the skeletal muscles of the trunk and limbs will form, are present at E8.5-9.5. Myogenin is a regulatory factor required for skeletal muscle differentiation. The data demonstrate that myogenin is associated with its own promoter in E8.5 and E9.5 embryos. Because myogenin is only expressed in somites at this stage of development, the data indicate that myogenin interactions with its own promoter have already occurred in skeletal muscle precursor cells in E8.5 embryos.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Regulación de la Expresión Génica , Análisis por Matrices de Proteínas/métodos , Animales , Cromatina/química , Cromatina/genética , Cromatina/aislamiento & purificación , Embrión de Mamíferos , Femenino , Masculino , Ratones , Miogenina/biosíntesis , Miogenina/genética , Embarazo , Regiones Promotoras Genéticas , Somitos/embriología , Somitos/fisiología
9.
EMBO J ; 25(1): 129-38, 2006 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-16319921

RESUMEN

Notch1 specifically upregulates expression of the cytokine interferon-gamma in peripheral T cells through activation of NF-kappaB. However, how Notch mediates NF-kappaB activation remains unclear. Here, we examined the temporal relationship between Notch signaling and NF-kappaB induction during T-cell activation. NF-kappaB activation occurs within minutes of T-cell receptor (TCR) engagement and this activation is sustained for at least 48 h following TCR signaling. We used gamma-secretase inhibitor (GSI) to prevent the cleavage and subsequent activation of Notch family members. We demonstrate that GSI blocked the later, sustained NF-kappaB activation, but did not affect the initial activation of NF-kappaB. Using biochemical approaches, as well as confocal microscopy, we show that the intracellular domain of Notch1 (N1IC) directly interacts with NF-kappaB and competes with IkappaBalpha, leading to retention of NF-kappaB in the nucleus. Additionally, we show that N1IC can directly regulate IFN-gamma expression through complexes formed on the IFN-gamma promoter. Taken together, these data suggest that there are two 'waves' of NF-kappaB activation: an initial, Notch-independent phase, and a later, sustained activation of NF-kappaB, which is Notch dependent.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Proteínas I-kappa B/metabolismo , Interferón gamma/genética , FN-kappa B/metabolismo , Receptor Notch1/metabolismo , Animales , Anticuerpos/farmacología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Núcleo Celular/química , Citosol/química , Citosol/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa , FN-kappa B/análisis , Subunidad p50 de NF-kappa B/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-rel/genética , Transducción de Señal , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T/inmunología
10.
Nat Immunol ; 6(7): 680-8, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15991363

RESUMEN

Notch receptors are processed by gamma-secretase acting in synergy with T cell receptor signaling to sustain peripheral T cell activation. Activated CD4+ T cells differentiate into T helper type 1 (TH1) or TH2 subsets. Molecular cues directing TH1 differentiation include expression of the TH1-specific transcription factor T-bet, encoded by Tbx21. However, the regulation of Tbx21 remains incompletely defined. Here we report that Notch1 can directly regulate Tbx21 through complexes formed on the Tbx21 promoter. In vitro, gamma-secretase inhibitors extinguished expression of Notch, interferon-gamma and Tbx21 in TH1-polarized CD4+ cells, whereas ectopic expression of activated Notch1 restored Tbx21 transcription. In vivo, administration of gamma-secretase inhibitors substantially impeded TH1-mediated disease progression in the mouse experimental autoimmune encephalomyelitis model of multiple sclerosis. Thus, using gamma-secretase inhibitors to modulate Notch signaling may prove beneficial in treating TH1-mediated autoimmunity.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Endopeptidasas/inmunología , Inhibidores de Proteasas/farmacología , Receptores de Superficie Celular/antagonistas & inhibidores , Células TH1/inmunología , Factores de Transcripción/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide , Animales , Ácido Aspártico Endopeptidasas , Citocinas/inmunología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Hipersensibilidad Tardía/tratamiento farmacológico , Hipersensibilidad Tardía/inmunología , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Receptor Notch1 , Receptores de Superficie Celular/inmunología , Proteínas de Dominio T Box , Células TH1/efectos de los fármacos , Células TH1/enzimología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA