Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 7(1): 465, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28352109

RESUMEN

Differentiation of regulatory Treg (Treg) in the periphery is critical to control inflammatory processes. Although polarization of inducible Treg (iTreg) often occurs in an inflammatory environment, the effects exerted by inflammation on human iTreg differentiation have not been extensively studied. We observed that IL-1ß significantly reduced the frequency of FOXP3+ T cells under iTreg-polarizing conditions. Mechanistically, we show that IL-1ß activated mTORC1 and downstream upregulated hypoxia inducible factor-1 (HIF-1α) expression. Using specific inhibitors, we demonstrated that both steps were critical in the deleterious effect of IL-1ß on Treg differentiation. Chemical stabilization of HIF-1α by Dimethyloxalylglycine (DMOG) also significantly impaired iTreg differentiation. Interestingly, while IL-1ß-treated cells exhibited only minor changes in metabolism, DMOG treatment decreased iTreg mitochondrial respiration and increased their glycolytic capacity. In conclusion, exposure to inflammatory stimuli profoundly inhibits human Treg differentiation HIF-1α dependent, suggesting that targeting HIF-1α could be a strategy to foster iTreg differentiation in an inflammatory milieu. However, IL-1ß deleterious effect does not appear to be completely driven by metabolic changes. These data thus suggest that several mechanisms contribute to the regulation of iTreg differentiation, but the timing and respective requirement for each pathway vary depending on the milieu in which iTreg differentiate.


Asunto(s)
Diferenciación Celular , Factores de Transcripción Forkhead/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-1beta/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Aminoácidos Dicarboxílicos/metabolismo , Biomarcadores , Diferenciación Celular/efectos de los fármacos , Respiración de la Célula , Expresión Génica , Glucólisis , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inmunofenotipificación , Mitocondrias/genética , Mitocondrias/metabolismo , Fenotipo , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/metabolismo
2.
Sci Rep ; 6: 35521, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27762344

RESUMEN

The transcription factor Interferon Regulatory Factor 4 (IRF4) is essential for TH2 and TH17 cell formation and controls peripheral CD8+ T cell differentiation. We used Listeria monocytogenes infection to characterize the function of IRF4 in TH1 responses. IRF4-/- mice generated only marginal numbers of listeria-specific TH1 cells. After transfer into infected mice, IRF4-/- CD4+ T cells failed to differentiate into TH1 cells as indicated by reduced T-bet and IFN-γ expression, and showed limited proliferation. Activated IRF4-/- CD4+ T cells exhibited diminished uptake of the glucose analog 2-NBDG, limited oxidative phosphorylation and strongly reduced aerobic glycolysis. Insufficient metabolic adaptation contributed to the limited proliferation and TH1 differentiation of IRF4-/- CD4+ T cells. Our study identifies IRF4 as central regulator of TH1 responses and cellular metabolism. We propose that this function of IRF4 is fundamental for the initiation and maintenance of all TH cell responses.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Factores Reguladores del Interferón/metabolismo , Células TH1/metabolismo , Animales , Factores Reguladores del Interferón/genética , Interferón gamma/genética , Interferón gamma/metabolismo , Ratones , Ratones Noqueados , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
3.
Immunol Cell Biol ; 94(1): 109-13, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26150319

RESUMEN

The transcription factor hypoxia inducible factor-1α (HIF-1α) mediates the metabolic adaptation of cells to hypoxia and T-helper cell fate. However, HIF-1α regulation in CD4(+) T cells (T cells) remains elusive. Here we observed that depletion of oxygen (O2⩽2%) alone was not sufficient to induce HIF-1α expression in T cells. However, when hypoxic T cells were stimulated, HIF-1α was expressed and this was dependent on nuclear factor-κB- and nuclear factor of activated T cell (NFAT)-mediated transcriptional upregulation of Hif-1α mRNA. HIF-1α upregulation could be blocked by drugs inhibiting NF-κB, NFAT or mammalian target of rapamycin precluding CD4(+) T-cell stimulation or translation in T cells, as well as by blocking transcription. CD3, CD28, phorbol-12-myristat-13-acetat (PMA) or ionomycin-stimulated T cells did not express HIF-1α under normoxic conditions. In conclusion, regulation of HIF-1α expression in CD4(+) T cells in hypoxia gravely relies on its transcriptional upregulation and subsequent enhanced protein stabilization.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Transcripción Genética , Linfocitos T CD4-Positivos/inmunología , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Activación de Linfocitos/inmunología , Masculino , Estabilidad Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
J Leukoc Biol ; 96(2): 305-12, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24664971

RESUMEN

The central oxygen sensitive transcription factor HIF-1α has been implicated in the differentiation of n(T(reg)) and Th17 cells and to orchestrate metabolic changes of activated T cells. However, data on the functional relevance of HIF-1α and Hox, in general, for nT(reg)-suppressive activity and T cell function in primary human cells are still missing. Therefore, we analyzed the effect of Hox and HIF-1α on human T(res), n(Treg), and Th17 cells. Under Hox, nT(reg)-mediated suppression of T(res) proliferation, CD25 expression, and secretion of IFN-γ were significantly reduced, whereas expression levels of VEGF, TNF-α, and IL-10 were significantly increased. In contrast to observations in mice, Th17 lineage commitment, as determined by RORγt expression, was not affected by activation or inhibition of HIF-1α expression using DMOG or YC-1 treatment, respectively. Nevertheless, the secretion of IL-17A was increased by DMOG and reduced by YC-1 under Th17-skewing conditions in a dose- dependent manner. In conclusion, Hox and HIF-1α substantially influence human T cell-mediated immune responses by modulation of nT(reg)-suppressive function and IL-17A secretion by Th17 cells.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Antígenos CD4/inmunología , Citocinas/inmunología , Activadores de Enzimas/farmacología , Femenino , Humanos , Indazoles/farmacología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Masculino , Ratones , Linfocitos T Reguladores/citología , Células Th17/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA