Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 81(1): 98, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38386110

ABSTRACT

In hormone-responsive breast cancer cells, progesterone (P4) has been shown to act via its nuclear receptor (nPR), a ligand-activated transcription factor. A small fraction of progesterone receptor is palmitoylated and anchored to the cell membrane (mbPR) forming a complex with estrogen receptor alpha (ERα). Upon hormone exposure, either directly or via interaction with ERα, mbPR activates the SRC/RAS/ERK kinase pathway leading to phosphorylation of nPR by ERK. Kinase activation is essential for P4 gene regulation, as the ERK and MSK1 kinases are recruited by the nPR to its genomic binding sites and trigger chromatin remodeling. An interesting open question is whether activation of mbPR can result in gene regulation in the absence of ligand binding to intracellular progesterone receptor (iPR). This matter has been investigated in the past using P4 attached to serum albumin, but the attachment is leaky and albumin can be endocytosed and degraded, liberating P4. Here, we propose a more stringent approach to address this issue by ensuring attachment of P4 to the cell membrane via covalent binding to a stable phospholipid. This strategy identifies the actions of P4 independent from hormone binding to iPR. We found that a membrane-attached progestin can activate mbPR, the ERK signaling pathway leading to iPR phosphorylation, initial gene regulation and entry into the cell cycle, in the absence of detectable intracellular progestin.


Subject(s)
Neoplasms , Progesterone , Progesterone/pharmacology , Receptors, Progesterone/genetics , Estrogen Receptor alpha , Progestins/pharmacology , Ligands , Cell Membrane
2.
Nucleic Acids Res ; 49(22): 12716-12731, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34850111

ABSTRACT

Here, we report that in T47D breast cancer cells 50 pM progestin is sufficient to activate cell cycle entry and the progesterone gene expression program. At this concentration, equivalent to the progesterone blood levels found around the menopause, progesterone receptor (PR) binds only to 2800 genomic sites, which are accessible to ATAC cleavage prior to hormone exposure. These highly accessible sites (HAs) are surrounded by well-organized nucleosomes and exhibit breast enhancer features, including estrogen receptor alpha (ERα), higher FOXA1 and BRD4 (bromodomain containing 4) occupancy. Although HAs are enriched in RAD21 and CTCF, PR binding is the driving force for the most robust interactions with hormone-regulated genes. HAs show higher frequency of 3D contacts among themselves than with other PR binding sites, indicating colocalization in similar compartments. Gene regulation via HAs is independent of classical coregulators and ATP-activated remodelers, relying mainly on MAP kinase activation that enables PR nuclear engagement. HAs are also preferentially occupied by PR and ERα in breast cancer xenografts derived from MCF-7 cells as well as from patients, indicating their potential usefulness as targets for therapeutic intervention.


Subject(s)
Breast Neoplasms/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Progestins/physiology , Animals , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation , Chromatin , Estrogen Receptor alpha/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System , MCF-7 Cells , Mice , Promegestone/pharmacology , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
3.
Nat Commun ; 11(1): 3019, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32541649

ABSTRACT

Transcription factors (TFs) regulate target genes by specific interactions with DNA sequences. Detecting and understanding these interactions at the molecular level is of fundamental importance in biological and clinical contexts. Crosslinking mass spectrometry is a powerful tool to assist the structure prediction of protein complexes but has been limited to the study of protein-protein and protein-RNA interactions. Here, we present a femtosecond laser-induced crosslinking mass spectrometry (fliX-MS) workflow, which allows the mapping of protein-DNA contacts at single nucleotide and up to single amino acid resolution. Applied to recombinant histone octamers, NF1, and TBP in complex with DNA, our method is highly specific for the mapping of DNA binding domains. Identified crosslinks are in close agreement with previous biochemical data on DNA binding and mostly fit known complex structures. Applying fliX-MS to cells identifies several bona fide crosslinks on DNA binding domains, paving the way for future large scale ex vivo experiments.


Subject(s)
DNA/chemistry , Mass Spectrometry/methods , NFI Transcription Factors/chemistry , TATA-Box Binding Protein/chemistry , Transcription Factors/chemistry , Animals , DNA/genetics , DNA/metabolism , Humans , Lasers , Mass Spectrometry/instrumentation , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism , Protein Binding , Protein Domains , Swine , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Nucleic Acids Res ; 48(8): 4147-4160, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32103264

ABSTRACT

Linker histones H1 are principal chromatin components, whose contribution to the epigenetic regulation of chromatin structure and function is not fully understood. In metazoa, specific linker histones are expressed in the germline, with female-specific H1s being normally retained in the early-embryo. Embryonic H1s are present while the zygotic genome is transcriptionally silent and they are replaced by somatic variants upon activation, suggesting a contribution to transcriptional silencing. Here we directly address this question by ectopically expressing dBigH1 in Drosophila S2 cells, which lack dBigH1. We show that dBigH1 binds across chromatin, replaces somatic dH1 and reduces nucleosome repeat length (NRL). Concomitantly, dBigH1 expression down-regulates gene expression by impairing RNApol II binding and histone acetylation. These effects depend on the acidic N-terminal ED-domain of dBigH1 since a truncated form lacking this domain binds across chromatin and replaces dH1 like full-length dBigH1, but it does not affect NRL either transcription. In vitro reconstitution experiments using Drosophila preblastodermic embryo extracts corroborate these results. Altogether these results suggest that the negatively charged N-terminal tail of dBigH1 alters the functional state of active chromatin compromising transcription.


Subject(s)
Chromatin/metabolism , Drosophila Proteins/metabolism , Gene Silencing , Histones/metabolism , Animals , Cell Line , Down-Regulation , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/chemistry , Histone Code , Histones/chemistry , Protein Domains , RNA Polymerase II/metabolism
6.
Sci Rep ; 8(1): 10927, 2018 Jul 19.
Article in English | MEDLINE | ID: mdl-30026552

ABSTRACT

Chromatin is known to undergo extensive remodeling during nuclear reprogramming. However, the factors and mechanisms involved in this remodeling are still poorly understood and current experimental approaches to study it are not best suited for molecular and genetic analyses. Here we report on the use of Drosophila preblastodermic embryo extracts (DREX) in chromatin remodeling experiments. Our results show that incubation of somatic nuclei in DREX induces changes in chromatin organization similar to those associated with nuclear reprogramming, such as rapid binding of the germline specific linker histone dBigH1 variant to somatic chromatin, heterochromatin reorganization, changes in the epigenetic state of chromatin, and nuclear lamin disassembly. These results raise the possibility of using the powerful tools of Drosophila genetics for the analysis of chromatin changes associated with this essential process.


Subject(s)
Chromatin/metabolism , Drosophila/embryology , Embryo, Nonmammalian/chemistry , Acetylation , Animals , Cellular Reprogramming , Chromatin/genetics , Chromatin Assembly and Disassembly , Drosophila/genetics , Drosophila/metabolism , Epigenesis, Genetic , Histones/metabolism
7.
Nat Commun ; 9(1): 250, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29343827

ABSTRACT

With a diverse network of substrates, NUDIX hydrolases have emerged as a key family of nucleotide-metabolizing enzymes. NUDT5 (also called NUDIX5) has been implicated in ADP-ribose and 8-oxo-guanine metabolism and was recently identified as a rheostat of hormone-dependent gene regulation and proliferation in breast cancer cells. Here, we further elucidate the physiological relevance of known NUDT5 substrates and underscore the biological requirement for NUDT5 in gene regulation and proliferation of breast cancer cells. We confirm the involvement of NUDT5 in ADP-ribose metabolism and dissociate a relationship to oxidized nucleotide sanitation. Furthermore, we identify potent NUDT5 inhibitors, which are optimized to promote maximal NUDT5 cellular target engagement by CETSA. Lead compound, TH5427, blocks progestin-dependent, PAR-derived nuclear ATP synthesis and subsequent chromatin remodeling, gene regulation and proliferation in breast cancer cells. We herein present TH5427 as a promising, targeted inhibitor that can be used to further study NUDT5 activity and ADP-ribose metabolism.


Subject(s)
Enzyme Inhibitors/pharmacology , Progestins/metabolism , Pyrophosphatases/antagonists & inhibitors , Signal Transduction/drug effects , Adenosine Diphosphate Ribose/metabolism , Adenosine Triphosphate/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Female , HL-60 Cells , Humans , Molecular Structure , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , RNA Interference , Substrate Specificity
8.
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
9.
Cell Rep ; 3(6): 2021-32, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23770244

ABSTRACT

Polo-like kinase 1 (PLK1) is a key regulator of cell division and is overexpressed in many types of human cancers. Compared to its well-characterized role in mitosis, little is known about PLK1 functions in interphase. Here, we report that PLK1 mediates estrogen receptor (ER)-regulated gene transcription in human breast cancer cells. PLK1 interacts with ER and is recruited to ER cis-elements on chromatin. PLK1-coactivated genes included classical ER target genes such as Ps2, Wisp2, and Serpina3 and were enriched in developmental and tumor-suppressive functions. Performing large-scale phosphoproteomics of estradiol-treated MCF7 cells in the presence or absence of the specific PLK1 inhibitor BI2536, we identified several PLK1 end targets involved in transcription, including the histone H3K4 trimethylase MLL2, the function of which on ER target genes was impaired by PLK1 inhibition. Our results propose a mechanism for the tumor-suppressive role of PLK1 in mammals as an interphase transcriptional regulator.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Estrogen/genetics , Benzamides/pharmacology , Breast Neoplasms/enzymology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Female , Gene Expression Regulation, Neoplastic , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , MCF-7 Cells , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pteridines , Receptors, Estrogen/metabolism , Signal Transduction , Transcription, Genetic , Tumor Cells, Cultured , Polo-Like Kinase 1
10.
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
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
12.
Genes Dev ; 25(8): 845-62, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21447625

ABSTRACT

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.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/metabolism , Nuclear Proteins/metabolism , Promegestone/pharmacology , Repressor Proteins/metabolism , p300-CBP Transcription Factors/metabolism , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histones , Humans , Immunoprecipitation , Jumonji Domain-Containing Histone Demethylases/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , RNA Interference , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , p300-CBP Transcription Factors/genetics
13.
J Biol Chem ; 285(4): 2622-31, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19940123

ABSTRACT

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.


Subject(s)
Histones/metabolism , Mammary Tumor Virus, Mouse/genetics , NFI Transcription Factors/metabolism , Receptors, Progesterone/metabolism , Breast Neoplasms , Cell Line, Tumor , Chromatin/physiology , Female , Hormones/metabolism , Humans , Luciferases/genetics , Nucleosomes/physiology , Promoter Regions, Genetic/physiology , RNA, Small Interfering , Response Elements/physiology , Transcription, Genetic/physiology , Transcriptional Activation/physiology
14.
PLoS Genet ; 5(7): e1000567, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19609353

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Chromatin Assembly and Disassembly , Hormones/genetics , Promoter Regions, Genetic , Breast Neoplasms/pathology , Cell Line, Tumor , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins , Female , Humans , Regulatory Sequences, Nucleic Acid , p300-CBP Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...