Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 3595, 2022 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-35739121

RESUMO

Differentiation of multipotent stem cells into mature cells is fundamental for development and homeostasis of mammalian tissues, and requires the coordinated induction of lineage-specific transcriptional programs and cell cycle withdrawal. To understand the underlying regulatory mechanisms of this fundamental process, we investigated how the tissue-specific transcription factors, CEBPA and CEBPE, coordinate cell cycle exit and lineage-specification in vivo during granulocytic differentiation. We demonstrate that CEBPA promotes lineage-specification by launching an enhancer-primed differentiation program and direct activation of CEBPE expression. Subsequently, CEBPE confers promoter-driven cell cycle exit by sequential repression of MYC target gene expression at the G1/S transition and E2F-meditated G2/M gene expression, as well as by the up-regulation of Cdk1/2/4 inhibitors. Following cell cycle exit, CEBPE unleashes the CEBPA-primed differentiation program to generate mature granulocytes. These findings highlight how tissue-specific transcription factors coordinate cell cycle exit with differentiation through the use of distinct gene regulatory elements.


Assuntos
Regulação da Expressão Gênica , Fatores de Transcrição , Animais , Ciclo Celular , Diferenciação Celular/genética , Granulócitos/metabolismo , Mamíferos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Cell Rep ; 24(3): 766-780, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30021172

RESUMO

Hematopoietic stem cells (HSCs) are considered a heterogeneous cell population. To further resolve the HSC compartment, we characterized a retinoic acid (RA) reporter mouse line. Sub-fractionation of the HSC compartment in RA-CFP reporter mice demonstrated that RA-CFP-dim HSCs were largely non-proliferative and displayed superior engraftment potential in comparison with RA-CFP-bright HSCs. Gene expression analysis demonstrated higher expression of RA-target genes in RA-CFP-dim HSCs, in contrast to the RA-CFP reporter expression, but both RA-CFP-dim and RA-CFP-bright HSCs responded efficiently to RA in vitro. Single-cell RNA sequencing (RNA-seq) of >1,200 HSCs showed that differences in cell cycle activity constituted the main driver of transcriptional heterogeneity in HSCs. Moreover, further analysis of the single-cell RNA-seq data revealed that stochastic low-level expression of distinct lineage-affiliated transcriptional programs is a common feature of HSCs. Collectively, this work demonstrates the utility of the RA-CFP reporter line as a tool for the isolation of superior HSCs.


Assuntos
Compartimento Celular , Ciclo Celular/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Transcrição Gênica , Animais , Ciclo Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Genoma , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteínas Luminescentes/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transcriptoma/genética , Tretinoína/farmacologia
3.
Cell Rep ; 23(9): 2744-2757, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29847803

RESUMO

Transcription factors PU.1 and CEBPA are required for the proper coordination of enhancer activity during granulocytic-monocytic (GM) lineage differentiation to form myeloid cells. However, precisely how these factors control the chronology of enhancer establishment during differentiation is not known. Through integrated analyses of enhancer dynamics, transcription factor binding, and proximal gene expression during successive stages of murine GM-lineage differentiation, we unravel the distinct kinetics by which PU.1 and CEBPA coordinate GM enhancer activity. We find no evidence of a pioneering function of PU.1 during late GM-lineage differentiation. Instead, we delineate a set of enhancers that gain accessibility in a CEBPA-dependent manner, suggesting a pioneering function of CEBPA. Analyses of Cebpa null bone marrow demonstrate that CEBPA controls PU.1 levels and, unexpectedly, that the loss of CEBPA results in an early differentiation block. Taken together, our data provide insights into how PU.1 and CEBPA functionally interact to drive GM-lineage differentiation.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/deficiência , Diferenciação Celular/genética , Elementos Facilitadores Genéticos/genética , Células Mieloides/citologia , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Animais , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular , Linhagem da Célula , Cromatina/metabolismo , Feminino , Regulação da Expressão Gênica , Granulócitos/citologia , Granulócitos/metabolismo , Camundongos , Monócitos/citologia , Monócitos/metabolismo , Células Mieloides/metabolismo , Ligação Proteica
4.
Genes Dev ; 29(18): 1915-29, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26385962

RESUMO

The balance between self-renewal and differentiation is crucial for the maintenance of hematopoietic stem cells (HSCs). Whereas numerous gene regulatory factors have been shown to control HSC self-renewal or drive their differentiation, we have relatively few insights into transcription factors that serve to restrict HSC differentiation. In the present work, we identify ETS (E-twenty-six)-related gene (ERG) as a critical factor protecting HSCs from differentiation. Specifically, loss of Erg accelerates HSC differentiation by >20-fold, thus leading to rapid depletion of immunophenotypic and functional HSCs. Molecularly, we could demonstrate that ERG, in addition to promoting the expression of HSC self-renewal genes, also represses a group of MYC targets, thereby explaining why Erg loss closely mimics Myc overexpression. Consistently, the BET domain inhibitor CPI-203, known to repress Myc expression, confers a partial phenotypic rescue. In summary, ERG plays a critical role in coordinating the balance between self-renewal and differentiation of HSCs.


Assuntos
Diferenciação Celular/genética , Células-Tronco Hematopoéticas/citologia , Proteínas Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células da Medula Óssea/fisiologia , Adesão Celular/genética , Movimento Celular/genética , Transformação Celular Neoplásica/genética , Células Cultivadas , Deleção de Genes , Camundongos , Proteínas Oncogênicas/genética , Fatores de Transcrição/genética , Regulador Transcricional ERG
5.
PLoS One ; 9(1): e84728, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24404186

RESUMO

Ageing and cancer is often associated with altered T cell distributions and this phenomenon has been suggested to be the main driver in the development of immunosenescence. Memory phenotype PD-1+ CD4+ T cells accumulate with age and during leukemic development, and they might account for the attenuated T cell response in elderly or diseased individuals. The transcription factor C/EBPα has been suggested to be responsible for the accumulation as well as for the senescent features of these cells including impaired TCR signaling and decreased proliferation. Thus modulating the activity of C/EBPα could potentially target PD-1+ CD4+ T cells and consequently, impede the development of immunosenescence. To exploit this possibility we tested the importance of C/EBPα for the development of age-dependent PD-1+ CD4+ T cells as well as its role in the accumulation of PD-1+ CD4+ T cells during leukemic progression. In contrast to earlier suggestions, we find that loss of C/EBPα expression in the lymphoid compartment led to an increase of PD-1+ CD4+ T cells specifically in old mice, suggesting that C/EBPα repress the accumulation of these cells in elderly by inhibiting their proliferation. Furthermore, C/EBPα-deficiency in the lymphoid compartment had no effect on leukemic development and did not affect the accumulation of PD-1+ CD4+ T cells. Thus, in addition to contradict earlier suggestions of a role for C/EBPα in immunosenescence, these findings efficiently discard the potential of using C/EBPα as a target for the alleviation of ageing/cancer-associated immunosenescence.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Memória Imunológica , Receptor de Morte Celular Programada 1/metabolismo , Fatores Etários , Envelhecimento , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Senescência Celular , Progressão da Doença , Expressão Gênica , Leucemia/genética , Leucemia/imunologia , Leucemia/metabolismo , Leucemia/mortalidade , Ativação Linfocitária , Camundongos , Baço/imunologia , Baço/metabolismo
6.
J Exp Med ; 211(1): 5-13, 2014 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24367003

RESUMO

MLL-fusion proteins are potent inducers of oncogenic transformation, and their expression is considered to be the main oncogenic driving force in ∼10% of human acute myeloid leukemia (AML) patients. These oncogenic fusion proteins are responsible for the initiation of a downstream transcriptional program leading to the expression of factors such as MEIS1 and HOXA9, which in turn can replace MLL-fusion proteins in overexpression experiments. To what extent MLL fusion proteins act on their own during tumor initiation, or if they collaborate with other transcriptional regulators, is unclear. Here, we have compared gene expression profiles from human MLL-rearranged AML to normal progenitors and identified the myeloid tumor suppressor C/EBPα as a putative collaborator in MLL-rearranged AML. Interestingly, we find that deletion of Cebpa rendered murine hematopoietic progenitors completely resistant to MLL-ENL-induced leukemic transformation, whereas C/EBPα was dispensable in already established AMLs. Furthermore, we show that Cebpa-deficient granulocytic-monocytic progenitors were equally resistant to transformation and that C/EBPα collaborates with MLL-ENL in the induction of a transcriptional program, which is also apparent in human AML. Thus, our studies demonstrate a key role of C/EBPα in MLL fusion-driven transformation and find that it sharply demarcates tumor initiation and maintenance.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Transformação Celular Neoplásica/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Leucemia Mieloide Aguda/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Biologia Computacional , Primers do DNA/genética , Citometria de Fluxo , Deleção de Genes , Perfilação da Expressão Gênica , Histona-Lisina N-Metiltransferase , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Proteína Meis1 , Proteínas de Neoplasias/metabolismo , Reação em Cadeia da Polimerase
7.
Mol Cell Biol ; 26(3): 1028-37, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16428455

RESUMO

The C/EBPalpha transcription factor regulates growth and differentiation of several tissues during embryonic development. Several hypotheses as to how C/EBPalpha inhibits cellular growth in vivo have been derived, mainly from studies of tissue culture cells. In fetal liver it has been proposed that a short, centrally located, 15-amino-acid proline-histidine-rich region (PHR) of C/EBPalpha is responsible for the growth-inhibitory function of the protein through its ability to interact with CDK2 and CDK4, thereby inhibiting their activities. Homozygous Cebpa(DeltaPHR/DeltaPHR) (DeltaPHR) mice, carrying a modified cebpa allele lacking amino acids 180 to 194, were born at the Mendelian ratio, reached adulthood, and displayed no apparent adverse phenotypes. When fetal livers from the DeltaPHR mice were analyzed for their expression of cell cycle markers, bromodeoxyuridine incorporation, cyclin-dependent kinase 2 kinase activity, and global gene expression, we failed to detect any cell cycle or developmental differences between the DeltaPHR mice and their control littermates. These in vivo data demonstrate that any C/EBPalpha-mediated growth repression via the PHR as well as the basic region is dispensable for proper embryonic development of, and cell cycle control in, the liver. Surprisingly, control experiments performed in C/EBPalpha null fetal livers yielded similar results.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Histidina/química , Fígado/embriologia , Prolina/química , Adipócitos/citologia , Sequência de Aminoácidos , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/química , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Diferenciação Celular , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Histidina/genética , Humanos , Fígado/metabolismo , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Prolina/genética , Estrutura Terciária de Proteína , Ratos , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA