Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
EMBO J ; 35(16): 1822-43, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27390128

ABSTRACT

Eukaryotic gene regulation is associated with changes in chromatin compaction that modulate access to DNA regulatory sequences relevant for transcriptional activation or repression. Although much is known about the mechanism of chromatin remodeling in hormonal gene activation, how repression is accomplished is much less understood. Here we report that in breast cancer cells, ligand-activated progesterone receptor (PR) is directly recruited to transcriptionally repressed genes involved in cell proliferation along with the kinases ERK1/2 and MSK1. PR recruits BRG1 associated with the HP1γ-LSD1 complex repressor complex, which is further anchored via binding of HP1γ to the H3K9me3 signal deposited by SUV39H2. In contrast to what is observed during gene activation, only BRG1 and not the BAF complex is recruited to repressed promoters, likely due to local enrichment of the pioneer factor FOXA1. BRG1 participates in gene repression by interacting with H1.2, facilitating its deposition and stabilizing nucleosome positioning around the transcription start site. Our results uncover a mechanism of hormone-dependent transcriptional repression and a novel role for BRG1 in progestin regulation of breast cancer cell growth.


Subject(s)
DNA Helicases/metabolism , DNA/metabolism , Gene Expression Regulation , Histones/metabolism , Hormones/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Cell Line, Tumor , Humans , Protein Binding
2.
Science ; 352(6290): 1221-5, 2016 Jun 03.
Article in English | MEDLINE | ID: mdl-27257257

ABSTRACT

Key nuclear processes in eukaryotes, including DNA replication, repair, and gene regulation, require extensive chromatin remodeling catalyzed by energy-consuming enzymes. It remains unclear how the ATP demands of such processes are met in response to rapid stimuli. We analyzed this question in the context of the massive gene regulation changes induced by progestins in breast cancer cells and found that ATP is generated in the cell nucleus via the hydrolysis of poly(ADP-ribose) to ADP-ribose. In the presence of pyrophosphate, ADP-ribose is used by the pyrophosphatase NUDIX5 to generate nuclear ATP. The nuclear source of ATP is essential for hormone-induced chromatin remodeling, transcriptional regulation, and cell proliferation.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Adenosine Triphosphate/biosynthesis , Cell Nucleus/metabolism , Chromatin Assembly and Disassembly , Progestins/metabolism , Pyrophosphatases/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Nucleus/drug effects , Cell Proliferation , Crystallography, X-Ray , Diphosphates/metabolism , Energy Metabolism , Female , Gene Expression Regulation , Humans , Hydrolysis , MCF-7 Cells , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Progestins/pharmacology , Protein Multimerization , Pyrophosphatases/chemistry , Pyrophosphatases/genetics
4.
PLoS Genet ; 11(4): e1005174, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25894978

ABSTRACT

While the importance of gene enhancers in transcriptional regulation is well established, the mechanisms and the protein factors that determine enhancers activity have only recently begun to be unravelled. Recent studies have shown that progesterone receptor (PR) binds regions that display typical features of gene enhancers. Here, we show by ChIP-seq experiments that the chromatin remodeler CHD8 mostly binds promoters under proliferation conditions. However, upon progestin stimulation, CHD8 re-localizes to PR enhancers also enriched in p300 and H3K4me1. Consistently, CHD8 depletion severely impairs progestin-dependent gene regulation. CHD8 binding is PR-dependent but independent of the pioneering factor FOXA1. The SWI/SNF chromatin-remodelling complex is required for PR-dependent gene activation. Interestingly, we show that CHD8 interacts with the SWI/SNF complex and that depletion of BRG1 and BRM, the ATPases of SWI/SNF complex, impairs CHD8 recruitment. We also show that CHD8 is not required for H3K27 acetylation, but contributes to increase accessibility of the enhancer to DNaseI. Furthermore, CHD8 was required for RNAPII recruiting to the enhancers and for transcription of enhancer-derived RNAs (eRNAs). Taken together our data demonstrate that CHD8 is involved in late stages of PR enhancers activation.


Subject(s)
DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Receptors, Progesterone/genetics , Transcription Factors/genetics , Transcription, Genetic , Acetylation , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Promoter Regions, Genetic , Protein Binding , RNA Polymerase II/genetics , Receptors, Progesterone/metabolism , Transcription Factors/metabolism
5.
RNA ; 21(3): 360-74, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25589247

ABSTRACT

Splicing of mRNA precursors can occur cotranscriptionally and it has been proposed that chromatin structure influences splice site recognition and regulation. Here we have systematically explored potential links between nucleosome positioning and alternative splicing regulation upon progesterone stimulation of breast cancer cells. We confirm preferential nucleosome positioning in exons and report four distinct profiles of nucleosome density around alternatively spliced exons, with RNA polymerase II accumulation closely following nucleosome positioning. Hormone stimulation induces switches between profile classes, correlating with a subset of alternative splicing changes. Hormone-induced exon inclusion often correlates with higher nucleosome occupancy at the exon or the preceding intronic region and with higher RNA polymerase II accumulation. In contrast, exons skipped upon hormone stimulation display low nucleosome densities even before hormone treatment, suggesting that chromatin structure primes alternative splicing regulation. Skipped exons frequently harbor binding sites for hnRNP AB, a hormone-induced splicing regulator whose knock down prevents some hormone-induced skipping events. Collectively, our results argue that a variety of chromatin architecture mechanisms can influence alternative splicing decisions.


Subject(s)
Alternative Splicing/genetics , Breast Neoplasms/genetics , Nucleosomes/genetics , Progesterone/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Exons/genetics , Female , Humans , Introns/genetics , Nucleosomes/metabolism , Progesterone/genetics , RNA Polymerase II/genetics , RNA Splice Sites/genetics , RNA, Messenger/genetics
6.
Genes Dev ; 28(19): 2151-62, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25274727

ABSTRACT

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.


Subject(s)
Chromatin/drug effects , Gene Expression Regulation/drug effects , Progestins/pharmacology , Cell Line, Tumor , Chromatin/chemistry , Chromatin Assembly and Disassembly/drug effects , Hormones/pharmacology , Humans
7.
Methods Mol Biol ; 1204: 1-14, 2014.
Article in English | MEDLINE | ID: mdl-25182756

ABSTRACT

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.


Subject(s)
Chromatin/metabolism , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Chromatin/genetics , Chromatin Immunoprecipitation/methods , Female , Gene Expression Regulation, Neoplastic , Genome Components , Humans
8.
Nucleic Acids Res ; 41(12): 6072-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23640331

ABSTRACT

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.


Subject(s)
RNA Polymerase II/metabolism , Receptors, Progesterone/metabolism , Transcription Elongation, Genetic , bcl-X Protein/genetics , Alternative Splicing , Binding Sites , CREB-Binding Protein/metabolism , Cell Line, Tumor , Chromatin/chemistry , Humans , Positive Transcriptional Elongation Factor B/metabolism , Promegestone/pharmacology , bcl-X Protein/biosynthesis , bcl-X Protein/metabolism , p300-CBP Transcription Factors/metabolism
9.
Genes Dev ; 27(10): 1179-97, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23699411

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Multiprotein Complexes/metabolism , Progesterone/metabolism , RNA, Long Noncoding/metabolism , Receptors, Progesterone/metabolism , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing/drug effects , Genome, Human/genetics , Histone Demethylases/metabolism , Histones/metabolism , Humans , Ligands , Mammary Tumor Virus, Mouse/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/drug effects , Phosphorylation , Progesterone/pharmacology , RNA, Long Noncoding/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
10.
Mol Cell ; 49(1): 67-79, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23177737

ABSTRACT

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.


Subject(s)
Nucleosomes/metabolism , Receptors, Progesterone/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Base Sequence , Binding Sites , Cell Line, Tumor , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Chromosome Mapping , Consensus Sequence , High-Throughput Nucleotide Sequencing , Histones/metabolism , Humans , Nucleosomes/physiology , Progestins/physiology , Protein Binding , Response Elements , Sequence Analysis, DNA
11.
Genes Dev ; 26(17): 1972-83, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22948662

ABSTRACT

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.


Subject(s)
Breast Neoplasms/enzymology , Cyclin-Dependent Kinase 2/metabolism , Gene Expression Regulation, Neoplastic , Poly(ADP-ribose) Polymerases/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Models, Molecular , Phosphorylation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Progestins/pharmacology , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL