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.
Sci Adv ; 10(9): eadj5107, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38427725

RESUMO

Cell fate decisions are achieved with gene expression changes driven by lineage-specific transcription factors (TFs). These TFs depend on chromatin remodelers including the Brahma-related gene 1 (BRG1)-associated factor (BAF) complex to activate target genes. BAF complex subunits are essential for development and frequently mutated in cancer. Thus, interrogating how BAF complexes contribute to cell fate decisions is critical for human health. We examined the requirement for the catalytic BAF subunit BRG1 in neural progenitor cell (NPC) specification from human embryonic stem cells. During the earliest stages of differentiation, BRG1 was required to establish chromatin accessibility at neuroectoderm-specific enhancers. Depletion of BRG1 dorsalized NPCs and promoted precocious neural crest specification and enhanced neuronal differentiation. These findings demonstrate that BRG1 mediates NPC specification by ensuring proper expression of lineage-specific TFs and appropriate activation of their transcriptional programs.


Assuntos
Cromatina , Placa Neural , Humanos , Cromatina/genética , DNA Helicases/genética , DNA Helicases/metabolismo , Placa Neural/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo
2.
Mol Cell ; 82(4): 803-815.e5, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35077705

RESUMO

The hormone-stimulated glucocorticoid receptor (GR) modulates transcription by interacting with thousands of enhancers and GR binding sites (GBSs) throughout the genome. Here, we examined the effects of GR binding on enhancer dynamics and investigated the contributions of individual GBSs to the hormone response. Hormone treatment resulted in genome-wide reorganization of the enhancer landscape in breast cancer cells. Upstream of the DDIT4 oncogene, GR bound to four sites constituting a hormone-dependent super enhancer. Three GBSs were required as hormone-dependent enhancers that differentially promoted histone acetylation, transcription frequency, and burst size. Conversely, the fourth site suppressed transcription and hormone treatment alleviated this suppression. GR binding within the super enhancer promoted a loop-switching mechanism that allowed interaction of the DDIT4 TSS with the active GBSs. The unique functions of each GR binding site contribute to hormone-induced transcriptional heterogeneity and demonstrate the potential for targeted modulation of oncogene expression.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Dexametasona/farmacologia , Elementos Facilitadores Genéticos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Receptores de Glucocorticoides/agonistas , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Sítios de Ligação , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética
3.
Commun Biol ; 3(1): 126, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32170217

RESUMO

Steroid hormone receptors such as the Glucocorticoid Receptor (GR) mediate transcriptional responses to hormones and are frequently targeted in the treatment of human diseases. Experiments using bulk populations of cells have provided a detailed picture of the global transcriptional hormone response but are unable to interrogate cell-to-cell transcriptional heterogeneity. To examine the glucocorticoid response in individual cells, we performed single cell RNA sequencing (scRNAseq) in a human breast cancer cell line. The transcriptional response to hormone was robustly detected in individual cells and scRNAseq provided additional statistical power to identify over 100 GR-regulated genes that were not detected in bulk RNAseq. scRNAseq revealed striking cell-to-cell variability in the hormone response. On average, individual hormone-treated cells showed a response at only 30% of the total set of GR target genes. Understanding the basis of this heterogeneity will be critical for the development of more precise models of steroid hormone signaling.


Assuntos
Neoplasias da Mama/genética , Dexametasona/farmacologia , Heterogeneidade Genética/efeitos dos fármacos , Glucocorticoides/farmacologia , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Transcriptoma/efeitos dos fármacos , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Receptores de Glucocorticoides/genética , Transcrição Gênica/efeitos dos fármacos
4.
Curr Opin Endocr Metab Res ; 15: 8-14, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35128145

RESUMO

Breast cancers are a diverse group of diseases and are often characterized by their expression of receptors for hormones such as estrogen and progesterone. Recently another steroid hormone receptor, the glucocorticoid receptor (GR) has been shown to be a key player in breast cancer progression, metastasis, and treatment. These receptors bind to chromatin to elicit transcriptional changes within cells, which are often inhibited by the structure of chromatin itself. Chromatin remodeling proteins, such as Brahma-related gene 1 (BRG1), function to overcome this physical inhibition of transcription factor function and have been linked to many cancers including breast cancer. Recent efforts to understand the interactions of BRG1 and GR, including genomic and single cell analyses, within breast cancers may give insight into personalized medicine and other potential treatments.

5.
Elife ; 72018 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-29792595

RESUMO

The Glucocorticoid Receptor (GR) alters transcriptional activity in response to hormones by interacting with chromatin at GR binding sites (GBSs) throughout the genome. Our work in human breast cancer cells identifies three classes of GBSs with distinct epigenetic characteristics and reveals that BRG1 interacts with GBSs prior to hormone exposure. The GBSs pre-occupied by BRG1 are more accessible and transcriptionally active than other GBSs. BRG1 is required for a proper and robust transcriptional hormone response and knockdown of BRG1 blocks recruitment of the pioneer factors FOXA1 and GATA3 to GBSs. Finally, GR interaction with FOXA1 and GATA3 binding sites was restricted to sites pre-bound by BRG1. These findings demonstrate that BRG1 establishes specialized chromatin environments that define multiple classes of GBS. This in turn predicts that GR and other transcriptional activators function via multiple distinct chromatin-based mechanisms to modulate the transcriptional response.


Assuntos
Cromatina/metabolismo , DNA Helicases/metabolismo , Regulação da Expressão Gênica , Proteínas Nucleares/metabolismo , Receptores de Glucocorticoides/metabolismo , Fatores de Transcrição/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Glucocorticoides/metabolismo , Humanos , Ligação Proteica , Transdução de Sinais
6.
PLoS Genet ; 12(8): e1006224, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27487356

RESUMO

Antisense transcription is a prevalent feature at mammalian promoters. Previous studies have primarily focused on antisense transcription initiating upstream of genes. Here, we characterize promoter-proximal antisense transcription downstream of gene transcription starts sites in human breast cancer cells, investigating the genomic context of downstream antisense transcription. We find extensive correlations between antisense transcription and features associated with the chromatin environment at gene promoters. Antisense transcription downstream of promoters is widespread, with antisense transcription initiation observed within 2 kb of 28% of gene transcription start sites. Antisense transcription initiates between nucleosomes regularly positioned downstream of these promoters. The nucleosomes between gene and downstream antisense transcription start sites carry histone modifications associated with active promoters, such as H3K4me3 and H3K27ac. This region is bound by chromatin remodeling and histone modifying complexes including SWI/SNF subunits and HDACs, suggesting that antisense transcription or resulting RNA transcripts contribute to the creation and maintenance of a promoter-associated chromatin environment. Downstream antisense transcription overlays additional regulatory features, such as transcription factor binding, DNA accessibility, and the downstream edge of promoter-associated CpG islands. These features suggest an important role for antisense transcription in the regulation of gene expression and the maintenance of a promoter-associated chromatin environment.


Assuntos
Elementos Antissenso (Genética)/genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Transcrição Gênica , Elementos Antissenso (Genética)/biossíntese , Cromatina/genética , Ilhas de CpG/genética , Regulação Fúngica da Expressão Gênica , Genômica , Código das Histonas/genética , Histonas/genética , Humanos , Proteínas Nucleares/biossíntese , Nucleossomos/genética , Ligação Proteica/genética , Alinhamento de Sequência
7.
Front Biosci ; 12: 3321-32, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17485302

RESUMO

Although "stem cell biology" is frequently described as a young field, the examination of pluripotency and its effects on embryonic cells has had an interesting and somewhat unusual history. After decades of research into the pluripotency of mammalian embryonic cells, the use of pluripotent cells came into prominence as mouse embryonic stem cells (ESC) provided the foundation of knockout mouse technology; however, the basic biology of pluripotency in embryonic cells was not extensively examined for roughly another twenty years until the creation of human embryonic stem cell lines. With the burgeoning potential of cell based therapies and roles of cancer stem cells in disease, understanding basic biological mechanisms regulating stem cell characteristics now presents great new opportunities. Therefore, it is not surprising that the underlying genetic and epigenetic forces allowing ESC to maintain pluripotency have been the focus of intense scientific scrutiny in recent years. In order to fully appreciate the importance of new discoveries regarding pluripotency in ESC, it is necessary to understand the role of pluripotency in normal embryonic development. The main purpose of this review is to highlight recent discoveries in the context of what was known about pluripotency and lineage commitment in the embryo prior to the bioinformatics and genomics age. In doing so we attempt to elucidate the importance and limitations of recent discoveries and identify important avenues for future research.


Assuntos
Células-Tronco Pluripotentes/citologia , Animais , Linhagem da Célula , Humanos , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA