Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Immunity ; 29(3): 423-37, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18799149

RESUMEN

Medullary thymic epithelial cells (mTECs) establish T cell self-tolerance through the expression of autoimmune regulator (Aire) and peripheral tissue-specific self-antigens. However, signals underlying mTEC development remain largely unclear. Here, we demonstrate crucial regulation of mTEC development by receptor activator of NF-kappaB (RANK) and CD40 signals. Whereas only RANK signaling was essential for mTEC development during embryogenesis, in postnatal mice, cooperation between CD40 and RANK signals was required for mTEC development to successfully establish the medullary microenvironment. Ligation of RANK or CD40 on fetal thymic stroma in vitro induced mTEC development in a tumor necrosis factor-associated factor 6 (TRAF6)-, NF-kappaB inducing kinase (NIK)-, and IkappaB kinase beta (IKKbeta)-dependent manner. These results show that developmental-stage-dependent cooperation between RANK and CD40 promotes mTEC development, thereby establishing self-tolerance.


Asunto(s)
Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Células Epiteliales/citología , Células Epiteliales/inmunología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Autotolerancia , Timo/citología , Animales , Autoinmunidad , Antígenos CD40/deficiencia , Diferenciación Celular , Células Epiteliales/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ligando RANK/deficiencia , Ligando RANK/metabolismo , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/metabolismo , Timo/embriología , Timo/inmunología , Timo/fisiología , Quinasa de Factor Nuclear kappa B
2.
Genes Cells ; 16(4): 437-47, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21401811

RESUMEN

Regulatory T cells (Tregs), a subset of CD4(+) helper T cells, are crucial for immunological self-tolerance. Defect in development or function of Tregs results in autoimmune disease in human and mice. Whereas it is known that Tregs mainly develop in the thymus, the molecular mechanism underlying development of Treg is not fully understood. TRAF6-deficient mice showed a severe defect in the Treg development in thymus. In vitro fetal thymic organ culture experiments indicated that the defect is ascribed to the absence of TRAF6 in thymic cells. Moreover, mixed fetal liver transfer experiments revealed that the development of Foxp3(+) cells differentiated from Traf6(-/-) hematopoietic cells was specifically impaired in the thymus, indicating cell-intrinsic requirement for TRAF6 in the Treg development. On the other hand, TRAF6 is not required for the development of conventional CD4(+) T cell. In addition, TGFß-dependent induction of Foxp3 in CD4(+) T cells in vitro was not impaired by the absence of TRAF6. Overall, our data indicate that TRAF6 plays an essential role on the commitment of immature thymocytes to thymic Tregs in cell-intrinsic fashion.


Asunto(s)
Linfocitos T Reguladores/fisiología , Factor 6 Asociado a Receptor de TNF/metabolismo , Timo/citología , Animales , Factores de Transcripción Forkhead/metabolismo , Técnicas In Vitro , Ratones , Linfocitos T Reguladores/citología , Timo/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
3.
Biochem Biophys Res Commun ; 414(4): 773-8, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-22005462

RESUMEN

NF-κB-inducing kinase (NIK) plays critical roles in the development of lymph nodes and Peyer's patches, and microarchitecture of the thymus and spleen via NF-κB activation. Alymphoplasia (aly/aly) mice have a point mutation in the NIK gene that causes a defect in the activation of an NF-κB member RelB. Here, we developed a novel method to determine the aly mutation by genetic typing using PCR. This method facilitated the easy establishment of a congeneic aly/aly mouse line. Indeed, we generated a mouse line with aly mutation on a BALB/cA background (BALB/cA-aly/aly). BALB/cA-aly/aly mice showed significant splenomegaly with extramedullary hemopoiesis, which was not significant in aly/aly mice on a C57BL/6 background. Interestingly, the splenomegaly and extramedullary hemopoiesis caused by the aly mutation was gender-dependent. These data together with previous reports on extramedullary hemopoiesis in RelB-deficient mice suggest that NIK-RelB signaling may be involved in the suppression of extramedullary hemopoiesis in adult mice.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Hematopoyesis Extramedular/genética , Proteínas Serina-Treonina Quinasas/genética , Factor de Transcripción ReIB/metabolismo , Animales , Secuencia de Bases , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutación , Bazo/fisiología , Esplenomegalia/genética , Factor de Transcripción ReIB/genética , Quinasa de Factor Nuclear kappa B
4.
Biochem Biophys Res Commun ; 408(4): 530-6, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21527253

RESUMEN

Medullary thymic epithelial cells (mTECs) are essential for thymic negative selection to prevent autoimmunity. Previous studies show that mTEC development is dependent on the signal transducers TRAF6 and NIK. However, the downstream target genes of signals controlled by these molecules remain unknown. We performed a microarray analysis on mRNAs down-regulated by deficiencies in TRAF6 or functional NIK in an in vitro organ culture of fetal thymic stromata (2DG-FTOC). An in silico analysis of transcription factor binding sites in plausible promoter regions of differentially expressed genes suggests that STAT1 is involved in TRAF6- and NIK-dependent gene expression. Indeed, the signal of RANK, a TNF receptor family member that activates TRAF6 and NIK, induces the activation of STAT1 in 2DG-FTOC. Moreover, RANK signaling induces the up-regulation of interferon (IFN)-stimulated gene (ISG) expression, suggesting that the RANKL-dependent activation of STAT1 up-regulates ISG expression. The RANKL-dependent expression levels of ISGs were reduced but not completely abolished in interferon α receptor 1-deficient (Ifnar1(-/-)) 2DG-FTOC. Our data suggest that RANK signaling induces ISG expression in both type I interferon-independent and interferon-dependent mechanisms.


Asunto(s)
Células Epiteliales/inmunología , Regulación de la Expresión Génica , Interferón Tipo I/inmunología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Autotolerancia/genética , Timo/inmunología , Animales , Feto , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor de Interferón alfa y beta/genética , Transducción de Señal , Células del Estroma/inmunología , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Quinasa de Factor Nuclear kappa B
5.
J Exp Med ; 213(8): 1441-58, 2016 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-27401343

RESUMEN

Medullary thymic epithelial cells (mTECs) expressing autoimmune regulator (Aire) are critical for preventing the onset of autoimmunity. However, the differentiation program of Aire-expressing mTECs (Aire(+) mTECs) is unclear. Here, we describe novel embryonic precursors of Aire(+) mTECs. We found the candidate precursors of Aire(+) mTECs (pMECs) by monitoring the expression of receptor activator of nuclear factor-κB (RANK), which is required for Aire(+) mTEC differentiation. pMECs unexpectedly expressed cortical TEC molecules in addition to the mTEC markers UEA-1 ligand and RANK and differentiated into mTECs in reaggregation thymic organ culture. Introduction of pMECs in the embryonic thymus permitted long-term maintenance of Aire(+) mTECs and efficiently suppressed the onset of autoimmunity induced by Aire(+) mTEC deficiency. Mechanistically, pMECs differentiated into Aire(+) mTECs by tumor necrosis factor receptor-associated factor 6-dependent RANK signaling. Moreover, nonclassical nuclear factor-κB activation triggered by RANK and lymphotoxin-ß receptor signaling promoted pMEC induction from progenitors exhibiting lower RANK expression and higher CD24 expression. Thus, our findings identified two novel stages in the differentiation program of Aire(+) mTECs.


Asunto(s)
Diferenciación Celular/inmunología , Células Epiteliales/inmunología , Regulación de la Expresión Génica/inmunología , Células Madre Embrionarias de Ratones/inmunología , Timo/inmunología , Factores de Transcripción/inmunología , Animales , Diferenciación Celular/genética , Células Epiteliales/citología , Regulación de la Expresión Génica/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología , Lectinas de Plantas/genética , Lectinas de Plantas/inmunología , Timo/citología , Factores de Transcripción/genética , Proteína AIRE
6.
J Exp Med ; 211(12): 2425-38, 2014 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-25385757

RESUMEN

Medullary thymic epithelial cells (mTECs) expressing the autoimmune regulator AIRE and various tissue-specific antigens (TSAs) are critical for preventing the onset of autoimmunity and may attenuate tumor immunity. However, molecular mechanisms controlling mTEC development remain elusive. Here, we describe the roles of the transcription factor Spi-B in mTEC development. Spi-B is rapidly up-regulated by receptor activator of NF-κB ligand (RANKL) cytokine signaling, which triggers mTEC differentiation, and in turn up-regulates CD80, CD86, some TSAs, and the natural inhibitor of RANKL signaling, osteoprotegerin (OPG). Spi-B-mediated OPG expression limits mTEC development in neonates but not in embryos, suggesting developmental stage-specific negative feedback regulation. OPG-mediated negative regulation attenuates cellularity of thymic regulatory T cells and tumor development in vivo. Hence, these data suggest that this negative RANKL-Spi-B-OPG feedback mechanism finely tunes mTEC development and function and may optimize the trade-off between prevention of autoimmunity and induction of antitumor immunity.


Asunto(s)
Células Epiteliales/inmunología , Tolerancia Inmunológica/inmunología , Proteínas Proto-Oncogénicas c-ets/inmunología , Timo/inmunología , Animales , Animales Recién Nacidos , Antígeno B7-1/inmunología , Antígeno B7-1/metabolismo , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Células Epiteliales/metabolismo , Retroalimentación Fisiológica , Femenino , Expresión Génica/inmunología , Tolerancia Inmunológica/genética , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/inmunología , Osteoprotegerina/metabolismo , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Ligando RANK/inmunología , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/inmunología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Timo/metabolismo , Quinasa de Factor Nuclear kappa B
7.
PLoS One ; 9(5): e95992, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24788966

RESUMEN

Virus-derived double-stranded RNAs (dsRNAs) are sensed in the cytosol by retinoic acid-inducible gene (RIG)-I-like receptors (RLRs). These induce the expression of type I IFN and proinflammatory cytokines through signaling pathways mediated by the mitochondrial antiviral signaling (MAVS) protein. TNF receptor-associated factor (TRAF) family proteins are reported to facilitate the RLR-dependent expression of type I IFN by interacting with MAVS. However, the precise regulatory mechanisms remain unclear. Here, we show the role of FK506-binding protein 51 (FKBP51) in regulating the dsRNA-dependent expression of type I IFN. The binding of FKBP51 to TRAF6 was first identified by "in vitro virus" selection and was subsequently confirmed with a coimmunoprecipitation assay in HEK293T cells. The TRAF-C domain of TRAF6 is required for its interaction, although FKBP51 does not contain the consensus motif for interaction with the TRAF-C domain. Besides TRAF6, we found that FKBP51 also interacts with TRAF3. The depletion of FKBP51 reduced the expression of type I IFN induced by dsRNA transfection or Newcastle disease virus infection in murine fibroblasts. Consistent with this, the FKBP51 depletion attenuated dsRNA-mediated phosphorylations of IRF3 and JNK and nuclear translocation of RelA. Interestingly, dsRNA stimulation promoted the accumulation of FKBP51 in the mitochondria. Moreover, the overexpression of FKBP51 inhibited RLR-dependent transcriptional activation, suggesting a scaffolding function for FKBP51 in the MAVS-mediated signaling pathway. Overall, we have demonstrated that FKBP51 interacts with TRAF proteins and facilitates the expression of type I IFN induced by cytosolic dsRNA. These findings suggest a novel role for FKBP51 in the innate immune response to viral infection.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Interferón Tipo I/genética , Mitocondrias/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Línea Celular , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Ratones , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Transporte de Proteínas , ARN Bicatenario/metabolismo , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Proteínas de Unión a Tacrolimus/genética
8.
J Immunol ; 179(10): 6799-807, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17982070

RESUMEN

Signal transduction pathways regulating NF-kappaB activation essential for microenvironment formation in secondary lymphoid organs remain to be determined. We investigated the effect of a deficiency of TNFR-associated factor 6 (TRAF6), which activates the classical NF-kappaB pathway, in splenic microenvironment formation. Two-week-old TRAF6-deficient mice showed severe defects in B cell follicle and marginal zone formation, similar to mutant mice defective in lymphotoxin (Lt) beta receptor (LtbetaR) signal induction of nonclassical NF-kappaB activation. However, analysis revealed a TRAF6 role in architecture formation distinct from its role in the early neonatal Lt signaling pathway. LtbetaR signal was essential for primary B cell cluster formation with initial differentiation of follicular dendritic cells (FDCs) in neonatal mice. In contrast, TRAF6 was dispensable for progression to this stage but was required for converting B cell clusters to B cell follicles and maintaining FDCs through to later stages. Fetal liver transfer experiments suggested that TRAF6 in radiation-resistant cells is responsible for follicle formation. Despite FDC-specific surface marker expression, FDCs in neonatal TRAF6-deficient mice had lost the capability to express CXCL13. These data suggest that developmentally regulated activation of TRAF6 in FDCs is required for inducing CXCL13 expression to maintain B cell follicles.


Asunto(s)
Quimiocina CXCL13/inmunología , Células Dendríticas Foliculares/inmunología , Linfotoxina beta/inmunología , Transducción de Señal/inmunología , Bazo/inmunología , Factor 6 Asociado a Receptor de TNF/inmunología , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/inmunología , Diferenciación Celular , Quimiocina CXCL13/biosíntesis , Células Dendríticas Foliculares/metabolismo , Hígado/crecimiento & desarrollo , Hígado/inmunología , Hígado/metabolismo , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/inmunología , Receptor beta de Linfotoxina/metabolismo , Linfotoxina beta/biosíntesis , Linfotoxina beta/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Mutantes , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Transducción de Señal/genética , Bazo/crecimiento & desarrollo , Bazo/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA