Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Cell Biol ; 25(12): 1833-1847, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37945904

RESUMO

MAF amplification increases the risk of breast cancer (BCa) metastasis through mechanisms that are still poorly understood yet have important clinical implications. Oestrogen-receptor-positive (ER+) BCa requires oestrogen for both growth and metastasis, albeit by ill-known mechanisms. Here we integrate proteomics, transcriptomics, epigenomics, chromatin accessibility and functional assays from human and syngeneic mouse BCa models to show that MAF directly interacts with oestrogen receptor alpha (ERα), thereby promoting a unique chromatin landscape that favours metastatic spread. We identify metastasis-promoting genes that are de novo licensed following oestrogen exposure in a MAF-dependent manner. The histone demethylase KDM1A is key to the epigenomic remodelling that facilitates the expression of the pro-metastatic MAF/oestrogen-driven gene expression program, and loss of KDM1A activity prevents this metastasis. We have thus determined that the molecular basis underlying MAF/oestrogen-mediated metastasis requires genetic, epigenetic and hormone signals from the systemic environment, which influence the ability of BCa cells to metastasize.


Assuntos
Neoplasias da Mama , Epigênese Genética , Receptor alfa de Estrogênio , Amplificação de Genes , Proteínas Proto-Oncogênicas c-maf , Animais , Feminino , Humanos , Camundongos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Cromatina , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Proteínas Proto-Oncogênicas c-maf/genética
2.
Mol Cell ; 83(15): 2673-2691.e7, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37506700

RESUMO

Cell cycle progression is linked to transcriptome dynamics and variations in the response of pluripotent cells to differentiation cues, mostly through unknown determinants. Here, we characterized the cell-cycle-associated transcriptome and proteome of mouse embryonic stem cells (mESCs) in naive ground state. We found that the thymine DNA glycosylase (TDG) is a cell-cycle-regulated co-factor of the tumor suppressor p53. Furthermore, TDG and p53 co-bind ESC-specific cis-regulatory elements and thereby control transcription of p53-dependent genes during self-renewal. We determined that the dynamic expression of TDG is required to promote the cell-cycle-associated transcriptional heterogeneity. Moreover, we demonstrated that transient depletion of TDG influences cell fate decisions during the early differentiation of mESCs. Our findings reveal an unanticipated role of TDG in promoting molecular heterogeneity during the cell cycle and highlight the central role of protein dynamics for the temporal control of cell fate during development.


Assuntos
Timina DNA Glicosilase , Proteína Supressora de Tumor p53 , Animais , Camundongos , Ciclo Celular/genética , Linhagem Celular , Regulação da Expressão Gênica , Timina DNA Glicosilase/genética , Timina DNA Glicosilase/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
Genes Dev ; 36(7-8): 451-467, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35450883

RESUMO

Genome organization plays a pivotal role in transcription, but how transcription factors (TFs) rewire the structure of the genome to initiate and maintain the programs that lead to oncogenic transformation remains poorly understood. Acute promyelocytic leukemia (APL) is a fatal subtype of leukemia driven by a chromosomal translocation between the promyelocytic leukemia (PML) and retinoic acid receptor α (RARα) genes. We used primary hematopoietic stem and progenitor cells (HSPCs) and leukemic blasts that express the fusion protein PML-RARα as a paradigm to temporally dissect the dynamic changes in the epigenome, transcriptome, and genome architecture induced during oncogenic transformation. We found that PML-RARα initiates a continuum of topologic alterations, including switches from A to B compartments, transcriptional repression, loss of active histone marks, and gain of repressive histone marks. Our multiomics-integrated analysis identifies Klf4 as an early down-regulated gene in PML-RARα-driven leukemogenesis. Furthermore, we characterized the dynamic alterations in the Klf4 cis-regulatory network during APL progression and demonstrated that ectopic Klf4 overexpression can suppress self-renewal and reverse the differentiation block induced by PML-RARα. Our study provides a comprehensive in vivo temporal dissection of the epigenomic and topological reprogramming induced by an oncogenic TF and illustrates how topological architecture can be used to identify new drivers of malignant transformation.


Assuntos
Leucemia Promielocítica Aguda , Diferenciação Celular/genética , Transformação Celular Neoplásica/genética , Humanos , Fator 4 Semelhante a Kruppel , Leucemia Promielocítica Aguda/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia
4.
Front Cell Dev Biol ; 9: 655201, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33996816

RESUMO

Polycomb group (PcG) of proteins are a group of highly conserved epigenetic regulators involved in many biological functions, such as embryonic development, cell proliferation, and adult stem cell determination. PHD finger protein 19 (PHF19) is an associated factor of Polycomb repressor complex 2 (PRC2), often upregulated in human cancers. In particular, myeloid leukemia cell lines show increased levels of PHF19, yet little is known about its function. Here, we have characterized the role of PHF19 in myeloid leukemia cells. We demonstrated that PHF19 depletion decreases cell proliferation and promotes chronic myeloid leukemia (CML) differentiation. Mechanistically, we have shown how PHF19 regulates the proliferation of CML through a direct regulation of the cell cycle inhibitor p21. Furthermore, we observed that MTF2, a PHF19 homolog, partially compensates for PHF19 depletion in a subset of target genes, instructing specific erythroid differentiation. Taken together, our results show that PHF19 is a key transcriptional regulator for cell fate determination and could be a potential therapeutic target for myeloid leukemia treatment.

5.
Sci Adv ; 6(43)2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33097530

RESUMO

Ewing sarcoma (EwS) is an aggressive tumor that affects adolescents and young adults. EwS is defined by a chromosomal translocation, EWSR1-FLI1 being the most common, that causes genome reprogramming through remodeling of enhancers. Here, we describe an unexpected function of RING1B, which is highly expressed in EwS. While retaining its repressive activity at Polycomb developmental regulated genes, RING1B colocalizes with EWSR1-FLI1 at active enhancers. We demonstrate that RING1B is necessary for the expression of key EWSR1-FLI1 targets by facilitating oncogene recruitment to their enhancers. Knockdown of RING1B impairs growth of tumor xenografts and expression of genes regulated by EWSR1-FLI1 bound enhancers. Pharmacological inhibition of AURKB with AZD1152 increases H2Aub levels causing down-regulation of RING1B/EWSR1-FLI1 common targets. Our findings demonstrate that RING1B is a critical modulator of EWSR1-FLI1-induced chromatin remodeling, and its inhibition is a potential therapeutic strategy for the treatment of these tumors.


Assuntos
Cromatina , Sarcoma de Ewing , Adolescente , Carcinogênese , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Cromatina/genética , Montagem e Desmontagem da Cromatina , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Fusão Oncogênica/genética , Proteína EWS de Ligação a RNA/genética , Sarcoma de Ewing/tratamento farmacológico , Sarcoma de Ewing/genética , Adulto Jovem
6.
Elife ; 92020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32155117

RESUMO

The Polycomb-like protein PHF19/PCL3 associates with PRC2 and mediates its recruitment to chromatin in embryonic stem cells. PHF19 is also overexpressed in many cancers. However, neither PHF19 targets nor misregulated pathways involving PHF19 are known. Here, we investigate the role of PHF19 in prostate cancer cells. We find that PHF19 interacts with PRC2 and binds to PRC2 targets on chromatin. PHF19 target genes are involved in proliferation, differentiation, angiogenesis, and extracellular matrix organization. Depletion of PHF19 triggers an increase in MTF2/PCL2 chromatin recruitment, with a genome-wide gain in PRC2 occupancy and H3K27me3 deposition. Transcriptome analysis shows that PHF19 loss promotes deregulation of key genes involved in growth, metastasis, invasion, and of factors that stimulate blood vessels formation. Consistent with this, PHF19 silencing reduces cell proliferation, while promotes invasive growth and angiogenesis. Our findings reveal a role for PHF19 in controlling the balance between cell proliferation and invasiveness in prostate cancer.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias da Próstata/metabolismo , Fatores de Transcrição/metabolismo , Fosfatos de Cálcio/metabolismo , Diferenciação Celular , Linhagem Celular , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Estudo de Associação Genômica Ampla , Humanos , Masculino , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Neoplasias da Próstata/genética , Fatores de Transcrição/genética
7.
Nat Genet ; 50(10): 1452-1462, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30224650

RESUMO

In embryonic stem cells (ESCs), developmental gene promoters are characterized by their bivalent chromatin state, with simultaneous modification by MLL2 and Polycomb complexes. Although essential for embryogenesis, bivalency is functionally not well understood. Here, we show that MLL2 plays a central role in ESC genome organization. We generate a catalog of bona fide bivalent genes in ESCs and demonstrate that loss of MLL2 leads to increased Polycomb occupancy. Consequently, promoters lose accessibility, long-range interactions are redistributed, and ESCs fail to differentiate. We pose that bivalency balances accessibility and long-range connectivity of promoters, allowing developmental gene expression to be properly modulated.


Assuntos
Diferenciação Celular/genética , Cromatina/genética , Cromatina/metabolismo , Histona-Lisina N-Metiltransferase/fisiologia , Células-Tronco Embrionárias Murinas/fisiologia , Proteína de Leucina Linfoide-Mieloide/fisiologia , Regiões Promotoras Genéticas , Animais , Células Cultivadas , Cromatina/química , Montagem e Desmontagem da Cromatina/genética , Drosophila , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Histona-Lisina N-Metiltransferase/genética , Camundongos , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas do Grupo Polycomb/metabolismo , Ligação Proteica/genética
8.
FEBS J ; 282(9): 1723-35, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25271128

RESUMO

The Polycomb group (PcG) of proteins form chromatin-binding complexes with histone-modifying activity. The two main PcG repressive complexes studied (PRC1 and PRC2) are generally associated with chromatin in its repressed state. PRC2 is responsible for methylation of histone H3 at lysine 27 (H3K27me3), an epigenetic mark that is linked with numerous biological processes, including development, adult homeostasis and cancer. The core canonical complex PRC2, which contains the EZH1/2, SUZ12 and EED proteins, may be extended and functionally manipulated through interactions with several other proteins. In this review, we focus on these PRC2-associated proteins. As PRC2 functions are diverse, the variability conferred by these sub-stoichiometrically associated members may help to understand specific changes in PRC2 activity, chromatin recruitment and distribution required for gene repression.


Assuntos
Doença , Proteínas do Grupo Polycomb/fisiologia , Células-Tronco/citologia , Animais , Humanos , Metilação , Proteínas do Grupo Polycomb/metabolismo , Células-Tronco/metabolismo
9.
Genes Dev ; 28(19): 2151-62, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25274727

RESUMO

The human genome is segmented into topologically associating domains (TADs), but the role of this conserved organization during transient changes in gene expression is not known. Here we describe the distribution of progestin-induced chromatin modifications and changes in transcriptional activity over TADs in T47D breast cancer cells. Using ChIP-seq (chromatin immunoprecipitation combined with high-throughput sequencing), Hi-C (chromosome capture followed by high-throughput sequencing), and three-dimensional (3D) modeling techniques, we found that the borders of the ∼ 2000 TADs in these cells are largely maintained after hormone treatment and that up to 20% of the TADs could be considered as discrete regulatory units where the majority of the genes are either transcriptionally activated or repressed in a coordinated fashion. The epigenetic signatures of the TADs are homogeneously modified by hormones in correlation with the transcriptional changes. Hormone-induced changes in gene activity and chromatin remodeling are accompanied by differential structural changes for activated and repressed TADs, as reflected by specific and opposite changes in the strength of intra-TAD interactions within responsive TADs. Indeed, 3D modeling of the Hi-C data suggested that the structure of TADs was modified upon treatment. The differential responses of TADs to progestins and estrogens suggest that TADs could function as "regulons" to enable spatially proximal genes to be coordinately transcribed in response to hormones.


Assuntos
Cromatina/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Progestinas/farmacologia , Linhagem Celular Tumoral , Cromatina/química , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Hormônios/farmacologia , Humanos
10.
Methods Mol Biol ; 1204: 1-14, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25182756

RESUMO

Understanding how eukaryotic gene regulation works implies unraveling the mechanisms used by transcription factors to access DNA information packaged in chromatin. The current view is that different cell types express different parts of the genome because they are equipped with different sets of transcription factors. A few transcription factors are called pioneer factors because they are able to bind to their sites in nucleosomes and to open up chromatin thus enabling access for other transcription factors, which are unable to recognize DNA packaged in nucleosomes. But it is also possible that the way DNA is organized in chromatin differs between cell types and contributes to cell identity by restricting or enhancing access to specific gene cohorts. To unravel these mechanisms we studied the interaction of progesterone receptor with the genome of breast cancer cells and found that it binds preferentially to sites organized in nucleosomes, which contribute to functional interactions leading to gene regulation.


Assuntos
Cromatina/metabolismo , Receptores de Progesterona/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Cromatina/genética , Imunoprecipitação da Cromatina/métodos , Feminino , Regulação Neoplásica da Expressão Gênica , Componentes Genômicos , Humanos
11.
PLoS One ; 9(5): e97311, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24859236

RESUMO

Although non-genomic steroid receptor pathways have been studied over the past decade, little is known about the direct gene expression changes that take place as a consequence of their activation. Progesterone controls proliferation of rat endometrial stromal cells during the peri-implantation phase of pregnancy. We showed that picomolar concentration of progestin R5020 mimics this control in UIII endometrial stromal cells via ERK1-2 and AKT activation mediated by interaction of Progesterone Receptor (PR) with Estrogen Receptor beta (ERb) and without transcriptional activity of endogenous PR and ER. Here we identify early downstream targets of cytoplasmic PR signaling and their possible role in endometrial stromal cell proliferation. Microarray analysis of global gene expression changes in UIII cells treated for 45 min with progestin identified 97 up- and 341 down-regulated genes. The most over-represented molecular functions were transcription factors and regulatory factors associated with cell proliferation and cell cycle, a large fraction of which were repressors down-regulated by hormone. Further analysis verified that progestins regulate Ccnd1, JunD, Usf1, Gfi1, Cyr61, and Cdkn1b through PR-mediated activation of ligand-free ER, ERK1-2 or AKT, in the absence of genomic PR binding. ChIP experiments show that progestin promoted the interaction of USF1 with the proximal promoter of the Cdc2 gene. Usf1 knockdown abolished Cdc2 progestin-dependent transcriptional regulation and cell proliferation, which also blocked Cdc2 knockdown. We conclude that progestin-induced proliferation of endometrial stromal cells is mediated by ERK1-2 and AKT dependent early regulation of USF1, which directly induces Cdc2. To our knowledge, this is the first description of early target genes of progestin-activated classical PR via crosstalk with protein kinases and independently of hormone receptor binding to the genomic targets.


Assuntos
Proteína Quinase CDC2/metabolismo , Cromatina/metabolismo , Endométrio/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Progestinas/farmacologia , Receptores de Progesterona/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Proteína de Ligação a CREB/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Redes Reguladoras de Genes/efeitos dos fármacos , Humanos , Promegestona/farmacologia , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Fatores Estimuladores Upstream/metabolismo
12.
Proc Natl Acad Sci U S A ; 111(13): 4892-7, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24639548

RESUMO

Estrogen receptor-α (ERα) has central role in hormone-dependent breast cancer and its ligand-induced functions have been extensively characterized. However, evidence exists that ERα has functions that are independent of ligands. In the present work, we investigated the binding of ERα to chromatin in the absence of ligands and its functions on gene regulation. We demonstrated that in MCF7 breast cancer cells unliganded ERα binds to more than 4,000 chromatin sites. Unexpectedly, although almost entirely comprised in the larger group of estrogen-induced binding sites, we found that unliganded-ERα binding is specifically linked to genes with developmental functions, compared with estrogen-induced binding. Moreover, we found that siRNA-mediated down-regulation of ERα in absence of estrogen is accompanied by changes in the expression levels of hundreds of coding and noncoding RNAs. Down-regulated mRNAs showed enrichment in genes related to epithelial cell growth and development. Stable ERα down-regulation using shRNA, which caused cell growth arrest, was accompanied by increased H3K27me3 at ERα binding sites. Finally, we found that FOXA1 and AP2γ binding to several sites is decreased upon ERα silencing, suggesting that unliganded ERα participates, together with other factors, in the maintenance of the luminal-specific cistrome in breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Receptor alfa de Estrogênio/metabolismo , Genoma Humano/genética , Sítios de Ligação , Neoplasias da Mama/patologia , Proliferação de Células , Imunoprecipitação da Cromatina , Feminino , Ontologia Genética , Humanos , Ligantes , Células MCF-7 , Reação em Cadeia da Polimerase , RNA Interferente Pequeno/metabolismo
13.
Nucleus ; 4(3): 189-94, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23756349

RESUMO

A major challenge of modern human biology is to understand how a differentiated somatic cell integrates the response to external signals in the complex context of basic metabolic and tissue-specific gene expression programs. This requires exploring two interconnected basic processes: the signaling network and the global function of the key transcription factors on which signaling acts to modulate gene expression. An apparently simple model to study these questions has been steroid hormones action, since their intracellular receptors both initiate signaling and are the key transcription factors orchestrating the cellular response. We have used progesterone action in breast cancer cells to elucidate the intricacies of progesterone receptor (PR) signaling crosstalk with protein kinases, histone modifying enzymes and ATP-dependent chromatin remodeling complexes. ( 1) Recently we have described the cistrome of PR in these cells at different times after addition of hormone and its relationship to chromatin structure. ( 2) The role of chromatin in transcription factor binding to the genome is still debated, but the dominant view is that factors bind preferentially to nucleosome-depleted regions, usually identified as DNaseI-hypersensitive sites (DHS). In contrast with this vision, we have shown that PR requires nucleosomes for optimal binding and function. In breast cancer cells treated with progestins we identified 25,000 PR binding sites (PRbs), the majority encompassing several copies of the hexanucleotide TGTYCY, highly abundant in the genome. We found that strong functional PRbs accumulate around progesterone-induced genes mainly in enhancers, are enriched in DHS but exhibit high nucleosome occupancy. Progestin stimulation results in remodeling of these nucleosomes with displacement of histones H1 and H2A/H2B dimers. Our results strongly suggest that nucleosomes play crucial role in PR binding and hormonal gene regulation.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/genética , Nucleossomos/efeitos dos fármacos , Progesterona/farmacologia , Receptores de Progesterona/genética , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Sítios de Ligação , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Histonas/metabolismo , Humanos , Nucleossomos/química , Nucleossomos/metabolismo , Progesterona/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Receptores de Progesterona/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo
14.
Nucleic Acids Res ; 41(12): 6072-86, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23640331

RESUMO

Steroid receptors were classically described for regulating transcription by binding to target gene promoters. However, genome-wide studies reveal that steroid receptors-binding sites are mainly located at intragenic regions. To determine the role of these sites, we examined the effect of progestins on the transcription of the bcl-x gene, where only intragenic progesterone receptor-binding sites (PRbs) were identified. We found that in response to hormone treatment, the PR is recruited to these sites along with two histone acetyltransferases CREB-binding protein (CBP) and GCN5, leading to an increase in histone H3 and H4 acetylation and to the binding of the SWI/SNF complex. Concomitant, a more relaxed chromatin was detected along bcl-x gene mainly in the regions surrounding the intragenic PRbs. PR also mediated the recruitment of the positive elongation factor pTEFb, favoring RNA polymerase II (Pol II) elongation activity. Together these events promoted the re-distribution of the active Pol II toward the 3'-end of the gene and a decrease in the ratio between proximal and distal transcription. These results suggest a novel mechanism by which PR regulates gene expression by facilitating the proper passage of the polymerase along hormone-dependent genes.


Assuntos
RNA Polimerase II/metabolismo , Receptores de Progesterona/metabolismo , Elongação da Transcrição Genética , Proteína bcl-X/genética , Processamento Alternativo , Sítios de Ligação , Proteína de Ligação a CREB/metabolismo , Linhagem Celular Tumoral , Cromatina/química , Humanos , Fator B de Elongação Transcricional Positiva/metabolismo , Promegestona/farmacologia , Proteína bcl-X/biossíntese , Proteína bcl-X/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
15.
Mol Cell ; 49(1): 67-79, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23177737

RESUMO

Elucidating the global function of a transcription factor implies the identification of its target genes and genomic binding sites. The role of chromatin in this context is unclear, but the dominant view is that factors bind preferentially to nucleosome-depleted regions identified as DNaseI-hypersensitive sites (DHS). Here we show by ChIP, MNase, and DNaseI assays followed by deep sequencing that the progesterone receptor (PR) requires nucleosomes for optimal binding and function. In breast cancer cells treated with progestins, we identified 25,000 PR binding sites (PRbs). The majority of these sites encompassed several copies of the hexanucleotide TGTYCY, which is highly abundant in the genome. We found that functional PRbs accumulate around progesterone-induced genes, mainly in enhancers. Most of these sites overlap with DHS but exhibit high nucleosome occupancy. Progestin stimulation results in remodeling of these nucleosomes with displacement of histones H1 and H2A/H2B dimers. Our results strongly suggest that nucleosomes are crucial for PR binding and hormonal gene regulation.


Assuntos
Nucleossomos/metabolismo , Receptores de Progesterona/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina , Imunoprecipitação da Cromatina , Mapeamento Cromossômico , Sequência Consenso , Sequenciamento de Nucleotídeos em Larga Escala , Histonas/metabolismo , Humanos , Nucleossomos/fisiologia , Progestinas/fisiologia , Ligação Proteica , Elementos de Resposta , Análise de Sequência de DNA
16.
Genes Dev ; 26(17): 1972-83, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22948662

RESUMO

Eukaryotic gene regulation implies that transcription factors gain access to genomic information via poorly understood processes involving activation and targeting of kinases, histone-modifying enzymes, and chromatin remodelers to chromatin. Here we report that progestin gene regulation in breast cancer cells requires a rapid and transient increase in poly-(ADP)-ribose (PAR), accompanied by a dramatic decrease of cellular NAD that could have broad implications in cell physiology. This rapid increase in nuclear PARylation is mediated by activation of PAR polymerase PARP-1 as a result of phosphorylation by cyclin-dependent kinase CDK2. Hormone-dependent phosphorylation of PARP-1 by CDK2, within the catalytic domain, enhances its enzymatic capabilities. Activated PARP-1 contributes to the displacement of histone H1 and is essential for regulation of the majority of hormone-responsive genes and for the effect of progestins on cell cycle progression. Both global chromatin immunoprecipitation (ChIP) coupled with deep sequencing (ChIP-seq) and gene expression analysis show a strong overlap between PARP-1 and CDK2. Thus, progestin gene regulation involves a novel signaling pathway that connects CDK2-dependent activation of PARP-1 with histone H1 displacement. Given the multiplicity of PARP targets, this new pathway could be used for the pharmacological management of breast cancer.


Assuntos
Neoplasias da Mama/enzimologia , Quinase 2 Dependente de Ciclina/metabolismo , Regulação Neoplásica da Expressão Gênica , Poli(ADP-Ribose) Polimerases/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Modelos Moleculares , Fosforilação , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/química , Poli(ADP-Ribose) Polimerases/genética , Progestinas/farmacologia , Estrutura Terciária de Proteína
17.
Cancer Cell ; 21(1): 5-7, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22264783

RESUMO

The mechanisms leading to the constitutive activation of NF-κB in cancers and the pathways upstream and downstream of this activation are not fully understood. In this issue of Cancer Cell, Yamagishi et al. demonstrate that Polycomb-mediated silencing of miR-31 is implicated in the aberrant activation of NF-κB signaling in tumors.

18.
Genes Dev ; 25(8): 845-62, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21447625

RESUMO

Gene regulation by external signals requires access of transcription factors to DNA sequences of target genes, which is limited by the compaction of DNA in chromatin. Although we have gained insight into how core histones and their modifications influence this process, the role of linker histones remains unclear. Here we show that, within the first minute of progesterone action, a complex cooperation between different enzymes acting on chromatin mediates histone H1 displacement as a requisite for gene induction and cell proliferation. First, activated progesterone receptor (PR) recruits the chromatin remodeling complexes NURF and ASCOM (ASC-2 [activating signal cointegrator-2] complex) to hormone target genes. The trimethylation of histone H3 at Lys 4 by the MLL2/MLL3 subunits of ASCOM, enhanced by the hormone-induced displacement of the H3K4 demethylase KDM5B, stabilizes NURF binding. NURF facilitates the PR-mediated recruitment of Cdk2/CyclinA, which is required for histone H1 displacement. Cooperation of ATP-dependent remodeling, histone methylation, and kinase activation, followed by H1 displacement, is a prerequisite for the subsequent displacement of histone H2A/H2B catalyzed by PCAF and BAF. Chromatin immunoprecipitation (ChIP) and sequencing (ChIP-seq) and expression arrays show that H1 displacement is required for hormone induction of most hormone target genes, some of which are involved in cell proliferation.


Assuntos
Histona Desmetilases com o Domínio Jumonji/metabolismo , Proteínas Nucleares/metabolismo , Promegestona/farmacologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Montagem e Desmontagem da Cromatina , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Histonas , Humanos , Imunoprecipitação , Histona Desmetilases com o Domínio Jumonji/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Interferência de RNA , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição de p300-CBP/genética
19.
Mol Endocrinol ; 24(11): 2088-98, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20484412

RESUMO

Steroid hormones regulate gene expression by interaction of their receptors with hormone-responsive elements on DNA or with other transcription factors, but they can also activate cytoplasmic signaling cascades. Rapid activation of Erk by progestins via an interaction of the progesterone receptor (PR) with the estrogen receptor is critical for transcriptional activation of the mouse mammary tumor virus (MMTV) promoter and other progesterone target genes. Erk activation leads to the phosphorylation of PR, activation of mitogen- and stress-activated protein kinase 1, and the recruitment of a complex of the three activated proteins and of P300/CBP-associated factor (PCAF) to a single nucleosome, resulting in the phosphoacetylation of histone H3 and the displacement of heterochromatin protein 1γ. Hormone-dependent gene expression requires ATP-dependent chromatin remodeling complexes. Two switch/sucrose nonfermentable-like complexes, Brahma-related gene 1-associated factor (BAF) and polybromo-BAF are present in breast cancer cells, but only BAF is recruited to the MMTV promoter and cooperates with PCAF during activation of hormone-responsive promoters. PCAF acetylates histone H3 at K14, an epigenetic mark recognized by BAF subunits, thus anchoring the complex to chromatin. BAF catalyzes localized displacement of histones H2A and H2B, facilitating access of nuclear factor 1 and additional PR complexes to the hidden hormone-responsive elements on the MMTV promoter. The linker histone H1 is a structural component of chromatin generally regarded as a general repressor of transcription. However, it contributes to a better regulation of the MMTV promoter by favoring a more homogeneous nucleosome positioning, thus reducing basal transcription and actually enhancing hormone induced transcription. During transcriptional activation, H1 is phosphorylated and displaced from the promoter. The kinase cyclin-dependent kinase 2 is activated after progesterone treatment and could catalyze progesterone-induced phosphorylation of histone H1 by chromatin remodeling complexes. The initial steps of gene induction by progestins involve changes in the chromatin organization of target promoters that require the activation of several kinase signaling pathways initiated by membrane anchored PR. Because these pathways also respond to other external signals, they serve to integrate the hormonal response in the global context of the cellular environment.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Progesterona/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Animais , Humanos , Modelos Biológicos
20.
Mol Endocrinol ; 23(6): 809-26, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19299443

RESUMO

Steroid hormone receptors act directly in the nucleus on the chromatin organization and transcriptional activity of several promoters. Furthermore, they have an indirect effect on cytoplasmic signal transduction pathways, including MAPK, impacting ultimately on gene expression. We are interested in distinguishing between the two modes of action of progesterone receptor (PR) on the control of gene expression and cell proliferation. For this, we have stably expressed, in PR-negative breast cancer cells, tagged forms of the PR isoform B mutated at regions involved either in DNA binding (DNA-binding domain) or in its ability to interact with the estrogen receptor and to activate the c-Src/MAPK/Erk/Msk cascade (estrogen receptor-interacting domain). Both mutants impair PR-mediated activation of a well-understood model promoter in response to progestin, as well as hormone-induced cell proliferation. Additional mutants affecting transactivation activity of PR (activation function 2) or a zinc-finger implicated in dimerization (D-box) have also been tested. Microarrays and gene expression experiments on these cell lines define the subsets of hormone-responsive genes regulated by different modes of action of PR isoform B, as well as genes in which the nuclear and nongenomic pathways cooperate. Correlation between CCND1 expression in the different cell lines and their ability to support cell proliferation confirms CCND1 as a key controller gene.


Assuntos
Regulação Neoplásica da Expressão Gênica , Receptores de Progesterona/química , Receptores de Progesterona/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Análise Mutacional de DNA , Ativação Enzimática/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Modelos Genéticos , Proteínas Mutantes/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Progestinas/farmacologia , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA