Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
EMBO J ; 43(9): 1770-1798, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38565950

RESUMEN

The cancer epigenome has been studied in cells cultured in two-dimensional (2D) monolayers, but recent studies highlight the impact of the extracellular matrix and the three-dimensional (3D) environment on multiple cellular functions. Here, we report the physical, biochemical, and genomic differences between T47D breast cancer cells cultured in 2D and as 3D spheroids. Cells within 3D spheroids exhibit a rounder nucleus with less accessible, more compacted chromatin, as well as altered expression of ~2000 genes, the majority of which become repressed. Hi-C analysis reveals that cells in 3D are enriched for regions belonging to the B compartment, have decreased chromatin-bound CTCF and increased fusion of topologically associating domains (TADs). Upregulation of the Hippo pathway in 3D spheroids results in the activation of the LATS1 kinase, which promotes phosphorylation and displacement of CTCF from DNA, thereby likely causing the observed TAD fusions. 3D cells show higher chromatin binding of progesterone receptor (PR), leading to an increase in the number of hormone-regulated genes. This effect is in part mediated by LATS1 activation, which favors cytoplasmic retention of YAP and CTCF removal.


Asunto(s)
Neoplasias de la Mama , Factor de Unión a CCCTC , Cromatina , Proteínas Serina-Treonina Quinasas , Humanos , Factor de Unión a CCCTC/metabolismo , Factor de Unión a CCCTC/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Cromatina/metabolismo , Cromatina/genética , Femenino , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Receptores de Progesterona/metabolismo , Receptores de Progesterona/genética , Vía de Señalización Hippo
2.
EMBO Rep ; 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890452

RESUMEN

Heterochromatin stability is crucial for progenitor proliferation during early neurogenesis. It relays on the maintenance of local hubs of H3K9me. However, understanding the formation of efficient localized levels of H3K9me remains limited. To address this question, we used neural stem cells to analyze the function of the H3K9me2 demethylase PHF2, which is crucial for progenitor proliferation. Through mass-spectroscopy and genome-wide assays, we show that PHF2 interacts with heterochromatin components and is enriched at pericentromeric heterochromatin (PcH) boundaries where it maintains transcriptional activity. This binding is essential for silencing the satellite repeats, preventing DNA damage and genome instability. PHF2's depletion increases the transcription of heterochromatic repeats, accompanied by a decrease in H3K9me3 levels and alterations in PcH organization. We further show that PHF2's PHD and catalytic domains are crucial for maintaining PcH stability, thereby safeguarding genome integrity. These results highlight the multifaceted nature of PHF2's functions in maintaining heterochromatin stability and regulating gene expression during neural development. Our study unravels the intricate relationship between heterochromatin stability and progenitor proliferation during mammalian neurogenesis.

3.
EMBO J ; 38(18): e101426, 2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31373033

RESUMEN

Steroid hormones are key gene regulators in breast cancer cells. While estrogens stimulate cell proliferation, progestins activate a single cell cycle followed by proliferation arrest. Here, we use biochemical and genome-wide approaches to show that progestins achieve this effect via a functional crosstalk with C/EBPα. Using ChIP-seq, we identify around 1,000 sites where C/EBPα binding precedes and helps binding of progesterone receptor (PR) in response to hormone. These regions exhibit epigenetic marks of active enhancers, and C/EBPα maintains an open chromatin conformation that facilitates loading of ligand-activated PR. Prior to hormone exposure, C/EBPα favors promoter-enhancer contacts that assure hormonal regulation of key genes involved in cell proliferation by facilitating binding of RAD21, YY1, and the Mediator complex. Knockdown of C/EBPα disrupts enhancer-promoter contacts and decreases the presence of these architectural proteins, highlighting its key role in 3D chromatin looping. Thus, C/EBPα fulfills a previously unknown function as a potential growth modulator in hormone-dependent breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Receptores de Progesterona/metabolismo , Animales , Neoplasias de la Mama/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Ratones , Trasplante de Neoplasias , Progestinas/farmacología , Regiones Promotoras Genéticas , Ensayos Antitumor por Modelo de Xenoinjerto , Factor de Transcripción YY1/metabolismo
4.
Genome Res ; 29(1): 29-39, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30552103

RESUMEN

In breast cancer cells, some topologically associating domains (TADs) behave as hormonal gene regulation units, within which gene transcription is coordinately regulated in response to steroid hormones. Here we further describe that responsive TADs contain 20- to 100-kb-long clusters of intermingled estrogen receptor (ESR1) and progesterone receptor (PGR) binding sites, hereafter called hormone-control regions (HCRs). In T47D cells, we identified more than 200 HCRs, which are frequently bound by unliganded ESR1 and PGR. These HCRs establish steady long-distance inter-TAD interactions between them and organize characteristic looping structures with promoters in their TADs even in the absence of hormones in ESR1+-PGR+ cells. This organization is dependent on the expression of the receptors and is further dynamically modulated in response to steroid hormones. HCRs function as platforms that integrate different signals, resulting in some cases in opposite transcriptional responses to estrogens or progestins. Altogether, these results suggest that steroid hormone receptors act not only as hormone-regulated sequence-specific transcription factors but also as local and global genome organizers.


Asunto(s)
Receptor alfa de Estrógeno/biosíntesis , Estrógenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Progesterona/farmacología , Receptores de Progesterona/biosíntesis , Elementos de Respuesta , Transducción de Señal/efectos de los fármacos , Receptor alfa de Estrógeno/genética , Humanos , Células MCF-7 , Receptores de Progesterona/genética
5.
Genes Dev ; 27(10): 1179-97, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23699411

RESUMEN

A close chromatin conformation precludes gene expression in eukaryotic cells. Genes activated by external cues have to overcome this repressive state by locally changing chromatin structure to a more open state. Although much is known about hormonal gene activation, how basal repression of regulated genes is targeted to the correct sites throughout the genome is not well understood. Here we report that in breast cancer cells, the unliganded progesterone receptor (PR) binds genomic sites and targets a repressive complex containing HP1γ (heterochromatin protein 1γ), LSD1 (lysine-specific demethylase 1), HDAC1/2, CoREST (corepressor for REST [RE1 {neuronal repressor element 1} silencing transcription factor]), KDM5B, and the RNA SRA (steroid receptor RNA activator) to 20% of hormone-inducible genes, keeping these genes silenced prior to hormone treatment. The complex is anchored via binding of HP1γ to H3K9me3 (histone H3 tails trimethylated on Lys 9). SRA interacts with PR, HP1γ, and LSD1, and its depletion compromises the loading of the repressive complex to target chromatin-promoting aberrant gene derepression. Upon hormonal treatment, the HP1γ-LSD1 complex is displaced from these constitutively poorly expressed genes as a result of rapid phosphorylation of histone H3 at Ser 10 mediated by MSK1, which is recruited to the target sites by the activated PR. Displacement of the repressive complex enables the loading of coactivators needed for chromatin remodeling and activation of this set of genes, including genes involved in apoptosis and cell proliferation. These results highlight the importance of the unliganded PR in hormonal regulation of breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Complejos Multiproteicos/metabolismo , Progesterona/metabolismo , ARN Largo no Codificante/metabolismo , Receptores de Progesterona/metabolismo , Sitios de Unión , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Cromatina/genética , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen/efectos de los fármacos , Genoma Humano/genética , Histona Demetilasas/metabolismo , Histonas/metabolismo , Humanos , Ligandos , Virus del Tumor Mamario del Ratón/genética , Complejos Multiproteicos/química , Complejos Multiproteicos/efectos de los fármacos , Fosforilación , Progesterona/farmacología , ARN Largo no Codificante/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
6.
Genes Dev ; 26(17): 1972-83, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22948662

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/enzimología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Modelos Moleculares , Fosforilación , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/química , Poli(ADP-Ribosa) Polimerasas/genética , Progestinas/farmacología , Estructura Terciaria de Proteína
7.
Genes Dev ; 25(8): 845-62, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21447625

RESUMEN

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.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas Nucleares/metabolismo , Promegestona/farmacología , Proteínas Represoras/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histonas , Humanos , Inmunoprecipitación , Histona Demetilasas con Dominio de Jumonji/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Interferencia de ARN , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción p300-CBP/genética
8.
Nucleic Acids Res ; 41(12): 6072-86, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23640331

RESUMEN

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.


Asunto(s)
ARN Polimerasa II/metabolismo , Receptores de Progesterona/metabolismo , Elongación de la Transcripción Genética , Proteína bcl-X/genética , Empalme Alternativo , Sitios de Unión , Proteína de Unión a CREB/metabolismo , Línea Celular Tumoral , Cromatina/química , Humanos , Factor B de Elongación Transcripcional Positiva/metabolismo , Promegestona/farmacología , Proteína bcl-X/biosíntesis , Proteína bcl-X/metabolismo , Factores de Transcripción p300-CBP/metabolismo
9.
PLoS Genet ; 5(7): e1000567, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19609353

RESUMEN

Steroid hormones regulate gene expression by interaction of their receptors with hormone responsive elements (HREs) and recruitment of kinases, chromatin remodeling complexes, and coregulators to their target promoters. Here we show that in breast cancer cells the BAF, but not the closely related PBAF complex, is required for progesterone induction of several target genes including MMTV, where it catalyzes localized displacement of histones H2A and H2B and subsequent NF1 binding. PCAF is also needed for induction of progesterone target genes and acetylates histone H3 at K14, an epigenetic mark that interacts with the BAF subunits by anchoring the complex to chromatin. In the absence of PCAF, full loading of target promoters with hormone receptors and BAF is precluded, and induction is compromised. Thus, activation of hormone-responsive promoters requires cooperation of at least two chromatin remodeling activities, BAF and PCAF.


Asunto(s)
Neoplasias de la Mama/genética , Ensamble y Desensamble de Cromatina , Hormonas/genética , Regiones Promotoras Genéticas , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN , Femenino , Humanos , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción p300-CBP
10.
J Biol Chem ; 285(4): 2622-31, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-19940123

RESUMEN

Steroid hormones induce transcription of their responsive genes by complex mechanisms including synergism between the hormone receptors and other transcription factors. On the mouse mammary tumor virus (MMTV) promoter progesterone induction is mediated by the reciprocal synergism between progesterone receptor (PR) and the ubiquitous transcription factor nuclear factor 1 (NF1). PR binding mediates ATP-dependent displacement of histone H2A and H2B, enabling NF1 access to its target site. In minichromosomes assembled in vitro NF1 binding facilitates access of PR to the hormone-responsive elements (HREs) by precluding reforming of the histone octamer, but the function of NF1 in living cells remains unclear. Here we show that depleting NF1 by small interfering RNAs or mutating the NF1-binding site significantly compromises transcription of the MMTV promoter. The central HREs 2 and 3 are not needed for ATP-dependent H2A/H2B displacement or NF1 binding but are critical for full PR binding and MMTV transactivation. We found that NF1 binding to the MMTV promoter on a H3/H4 histone tetramer particle exposes the central HREs and facilitates their binding by PR, suggesting a possible mechanism for the reciprocal synergism between PR and NF1.


Asunto(s)
Histonas/metabolismo , Virus del Tumor Mamario del Ratón/genética , Factores de Transcripción NFI/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias de la Mama , Línea Celular Tumoral , Cromatina/fisiología , Femenino , Hormonas/metabolismo , Humanos , Luciferasas/genética , Nucleosomas/fisiología , Regiones Promotoras Genéticas/fisiología , ARN Interferente Pequeño , Elementos de Respuesta/fisiología , Transcripción Genética/fisiología , Activación Transcripcional/fisiología
11.
J Steroid Biochem Mol Biol ; 109(3-5): 344-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18417338

RESUMEN

Gene regulation by steroid hormones involves genomic and non-genomic signaling pathways and the relationship between these two pathways is unknown. Genomic actions are often mediated by binding of the ligand-activated hormone receptors to hormone responsive elements (HREs) followed by recruitment of co-regulators, remodeling of chromatin and formation of the transcription initiation complex. The non-genomic effects of steroid hormones involve the rapid and transient activation of several kinase cascades often mediated by a subpopulation of "nuclear" receptors located in the cytoplasmic side of the cell membrane. The progesterone effect on breast cancer cell proliferation involves activation of the Src/Ras/Erk cascade mediated by a specific interaction between two domains of the N-terminal half of PR and the ligand-binding domain of ERalpha. Unexpectedly, selective inhibition of Erk, or its target kinase Msk1, interferes with chromatin remodeling and blocks MMTV transcriptional activation. A complex of activated PR, Erk and Msk1 is recruited to promoter already 5 min after hormone treatment and phosphorylates histone H3 at serine 10, leading to displacement of HP1gamma, as a requisite for recruitment of Src1, chromatin remodeling complexes (hSnf2h and Brg1) and RNA polymerase II. Thus, activation of signaling cascades in the cytoplasm is essential for chromatin remodeling and transcriptional activation of a subset of steroid hormone target genes.


Asunto(s)
Cromatina/genética , Genoma/genética , Hormonas/metabolismo , Transducción de Señal , Betaretrovirus/genética , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Histonas/metabolismo , Hormonas/farmacología , Humanos , Nucleosomas/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica , Receptores de Progesterona/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Transcription ; 8(1): 15-20, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27700223

RESUMEN

How genes are repressed by steroid hormones remains a matter of debate, and several indirect mechanisms have been proposed. We found that the ligand-activated progesterone receptor recruits to the promoter of downregulated genes a repressor complex composed of HP1γ, the lysine demethylase LSD1, histone deacetylases, coREST, the RNA SRA, and the ATPase BRG1. BRG1 is needed for chromatin remodeling and facilitates the deposition of linker histone variant H1.2, which compacts chromatin and hinders RNA polymerase loading and transcription. Thus, steroid hormone receptors can repress genes in ways reminiscent of those used for gene induction, namely by directly targeting factors that remodel chromatin. But while PR-dependent gene induction in T47D cells is mainly achieved by potentiating enhancer activity, repression acts at the level of gene promoters.


Asunto(s)
Cromatina/genética , Silenciador del Gen , Receptores de Esteroides/metabolismo , Animales , Línea Celular , Ensamble y Desensamble de Cromatina , Redes Reguladoras de Genes , Humanos , Regiones Promotoras Genéticas , Activación Transcripcional
13.
Methods Mol Biol ; 1204: 1-14, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25182756

RESUMEN

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.


Asunto(s)
Cromatina/metabolismo , Receptores de Progesterona/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular , Línea Celular Tumoral , Cromatina/genética , Inmunoprecipitación de Cromatina/métodos , Femenino , Regulación Neoplásica de la Expresión Génica , Componentes Genómicos , Humanos
14.
Mol Endocrinol ; 24(11): 2088-98, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20484412

RESUMEN

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.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Progesterona/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal , Animales , Humanos , Modelos Biológicos
15.
Nucl Recept Signal ; 7: e008, 2009 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-20087429

RESUMEN

Transcription from the mouse mammary tumor virus (MMTV) promoter can be induced by progestins. The progesterone receptor (PR) binds to a cluster of five hormone responsive elements (HREs) and activates the promoter by synergistic interactions with the ubiquitous transcription factor, nuclear factor 1 (NF1). Progesterone treatment of cells in culture leads to activation of the Src/Ras/Erk/Msk1 cascade. Selective inhibition of Erk, or its target kinase Msk1, interferes with chromatin remodeling and blocks MMTV activation. A complex of activated PR, Erk and Msk1 is recruited to promoter after 5 min of hormone treatment and phosphorylates histone H3 at serine 10. This modification promotes the displacement of HP1gamma and subsequent chromatin remodeling. Progestin treatment leads to the recruitment of the BAF complex, which selectively displaces histones H2A and H2B from the nucleosome containing the HREs. The acetyltransferase PCAF is also required for induction of progesterone target genes and acetylates histone H3 at K14, an epigenetic mark, which interacts with Brg1 and Brm, anchoring the BAF complex to chromatin. In nucleosomes assembled on either MMTV or mouse rDNA promoter sequences, SWI/SNF displaces histones H2A and H2B from MMTV, but not from the rDNA nucleosome. Thus, the outcome of nucleosome remodeling by purified SWI/SNF depends on DNA sequence. The resultant H3/H4 tetramer particle is then the substrate for subsequent events in induction. Thus, initial activation of the MMTV promoter requires activation of several kinases and PCAF leading to phosphoacetylation of H3, and recruitment of BAF with subsequent removal of H2A/H2B.


Asunto(s)
Cromatina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Genes Virales/efectos de los fármacos , Genes Virales/genética , Virus del Tumor Mamario del Ratón/efectos de los fármacos , Virus del Tumor Mamario del Ratón/genética , Progestinas/farmacología , Animales , Histonas/metabolismo , Humanos
16.
Mol Cell ; 24(3): 367-81, 2006 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-17081988

RESUMEN

How genes are regulated in the context of chromatin is a central question of biology. Steroid hormones control gene expression via interaction of their receptors with target sequences on DNA but can also activate cytoplasmic signaling cascades. Here we report that rapid Erk activation by progestins participates in induction of target genes by preparing the chromatin for transcription. Five minutes after hormone treatment, Erk activation leads to phosphorylation of the progesterone receptor (PR), activation of Msk1, and recruitment of a complex of the three proteins to a nucleosome on the MMTV promoter. Msk1 phosphorylates histone H3, leading to displacement of HP1gamma and recruitment of Brg1 and RNA polymerase II. Cell-free experiments show a direct interaction between PR, Erk, and Msk1 and support the importance of H3 phosphorylation for nucleosome remodeling. Inhibition of Msk1 activation blocks recruitment of the kinase complex, H3 phosphorylation, and HP1gamma displacement, thus precluding remodeling and induction of the promoter.


Asunto(s)
Ensamble y Desensamble de Cromatina/efectos de los fármacos , Histonas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Progestinas/farmacología , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Animales , Sistema Libre de Células , Proteínas Cromosómicas no Histona/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Lisina/metabolismo , Virus del Tumor Mamario del Ratón/genética , Metilación/efectos de los fármacos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Nucleosomas/efectos de los fármacos , Nucleosomas/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , ARN Polimerasa II/metabolismo , Receptores de Progesterona/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Serina/metabolismo , Especificidad por Sustrato , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
17.
Mol Cell ; 16(3): 439-52, 2004 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-15525516

RESUMEN

Regulation of gene expression requires dynamic changes in chromatin, but the nature of these changes is not well understood. Here, we show that progesterone treatment of cultured cells leads to recruitment of progesterone receptor (PR) and SWI/SNF-related complexes to Mouse Mammary Tumor Virus (MMTV) promoter, accompanied by displacement of histones H2A and H2B from the nucleosome containing the receptor binding sites, but not from adjacent nucleosomes. PR recruits SWI/SNF to MMTV nucleosomes in vitro and facilitates synergistic binding of receptors and nuclear factor 1 to the promoter. In nucleosomes assembled on MMTV or mouse rDNA promoter sequences, SWI/SNF catalyzes ATP-dependent sliding of the histone octamer followed only on the MMTV promoter by displacement of histones H2A and H2B. In MMTV nucleosome arrays, SWI/SNF displaces H2A and H2B from nucleosome B and not from the adjacent nucleosome. Thus, the outcome of nucleosome remodeling by SWI/SNF depends on DNA sequence.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Adenosina Trifosfato/metabolismo , Sitios de Unión , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Ribosómico/genética , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Histonas/genética , Humanos , Virus del Tumor Mamario del Ratón/genética , Factores de Transcripción NFI , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Progesterona/farmacología , Receptores de Progesterona/metabolismo , Factores de Transcripción/genética , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA