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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Immunol ; 44(8): 2478-88, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24810486

RESUMO

DCs develop from multipotent progenitors (MPPs), which commit into DC-restricted common dendritic cell progenitors (CDPs). CDPs further differentiate into classical DCs (cDCs) and plasmacytoid DCs (pDCs). Here, we studied the impact of histone acetylation on DC development in C57BL/6 mice by interfering with histone acetylation and deacetylation, employing histone deacetylase (HDAC) inhibitors. We observed that commitment of MPPs into CDPs was attenuated by HDAC inhibition and that pDC development was specifically blocked. Gene expression profiling revealed that HDAC inhibition prevents establishment of a DC-specific gene expression repertoire. Importantly, protein levels of the core DC transcription factor PU.1 were reduced in HDAC inhibitor-treated cells and consequently PU.1 recruitment at PU.1 target genes Fms-like tyrosine kinase 3 (Flt3), interferon regulatory factor 8 (IRF8), and PU.1 itself was impaired. Thus, our results demonstrate that attenuation of PU.1 expression by HDAC inhibition causes reduced expression of key DC regulators, which results in attenuation of DC development. We propose that chromatin modifiers, such as HDACs, are required for establishing a DC gene network, where Flt3/STAT3 signaling drives PU.1 and IRF8 expression and DC development. Taken together, our study identifies HDACs as critical regulators of DC lineage commitment and development.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/enzimologia , Histona Desacetilases/metabolismo , Acetilação , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Células Dendríticas/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Histona Desacetilases/genética , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/enzimologia , Células-Tronco/metabolismo , Transativadores/genética , Transativadores/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
2.
Oncoimmunology ; 2(3): e23083, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23687620

RESUMO

We have recently described two types of Langerhans cells (LCs), which develop via separate pathways in steady-state conditions and during inflammation. Here, we propose that these two types of LCs differ in their requirement for transforming growth factor ß1 (TGFß1), and we discuss how TGFß1 impacts on the development of other dendritic cell subtypes.

3.
Eur J Cell Biol ; 91(6-7): 515-23, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22078373

RESUMO

Dendritic cells (DC) develop from hematopoietic stem cells, which is guided by instructive signals through cytokines. DC development progresses from multipotent progenitors (MPP) via common DC progenitors (CDP) into DC. Flt3 ligand (Flt3L) signaling via the Flt3/Stat3 pathway is of pivotal importance for DC development under steady state conditions. Additional factors produced during steady state or inflammation, such as TGF-ß1 or GM-CSF, also influence the differentiation potential of MPP and CDP. Here, we studied how gp130, GM-CSF and TGF-ß1 signaling influence DC lineage commitment from MPP to CDP and further into DC. We observed that activation of gp130 signaling promotes expansion of MPP. Additionally, gp130 signaling inhibited Flt3L-driven DC differentiation, but had little effect on GM-CSF-driven DC development. The inflammatory cytokine GM-CSF induces differentiation of MPP into inflammatory DC and blocks steady state DC development. Global transcriptome analysis revealed a GM-CSF-driven gene expression repertoire that primes MPP for differentiation into inflammatory DC. Finally, TGF-ß1 induces expression of DC-lineage affiliated genes in MPP, including Flt3, Irf-4 and Irf-8. Under inflammatory conditions, however, the effect of TGF-ß1 is altered: Flt3 is not upregulated, indicating that an inflammatory environment inhibits steady state DC development. Altogether, our data indicate that distinct cytokine signals produced during steady state or inflammation have a different outcome on DC lineage commitment and differentiation.


Assuntos
Citocinas/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Hematopoese/imunologia , Animais , Diferenciação Celular , Linhagem da Célula , Citocinas/metabolismo , Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Camundongos , Camundongos Endogâmicos C57BL
4.
J Immunol ; 185(9): 5326-35, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20881193

RESUMO

Dendritic cells (DCs) in lymphoid tissue comprise conventional DCs (cDCs) and plasmacytoid DCs (pDCs) that develop from common DC progenitors (CDPs). CDPs are Flt3(+)c-kit(int)M-CSFR(+) and reside in bone marrow. In this study, we describe a two-step culture system that recapitulates DC development from c-kit(hi)Flt3(-/lo) multipotent progenitors (MPPs) into CDPs and further into cDC and pDC subsets. MPPs and CDPs are amplified in vitro with Flt3 ligand, stem cell factor, hyper-IL-6, and insulin-like growth factor-1. The four-factor mixture readily induces self-renewal of MPPs and their progression into CDPs and has no self-renewal activity on CDPs. The amplified CDPs respond to all known DC poietins and generate all lymphoid tissue DCs in vivo and in vitro. Additionally, in vitro CDPs recapitulate the cell surface marker and gene expression profile of in vivo CDPs and possess a DC-primed transcription profile. TGF-ß1 impacts on CDPs and directs their differentiation toward cDCs. Genome-wide gene expression profiling of TGF-ß1-induced genes identified instructive transcription factors for cDC subset specification, such as IFN regulatory factor-4 and RelB. TGF-ß1 also induced the transcription factor inhibitor of differentiation/DNA binding 2 that suppresses pDC development. Thus, TGF-ß1 directs CDP differentiation into cDCs by inducing both cDC instructive factors and pDC inhibitory factors.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células-Tronco Hematopoéticas/citologia , Fator de Crescimento Transformador beta1/imunologia , Animais , Separação Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Citometria de Fluxo , Expressão Gênica , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/imunologia , Células-Tronco Multipotentes/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Crescimento Transformador beta1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA