Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Struct Mol Biol ; 31(3): 513-522, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38196033

ABSTRACT

Mitotic bookmarking transcription factors (TFs) are thought to mediate rapid and accurate reactivation after mitotic gene silencing. However, the loss of individual bookmarking TFs often leads to the deregulation of only a small proportion of their mitotic targets, raising doubts on the biological significance and importance of their bookmarking function. Here we used targeted proteomics of the mitotic bookmarking TF ESRRB, an orphan nuclear receptor, to discover a large redundancy in mitotic binding among members of the protein super-family of nuclear receptors. Focusing on the nuclear receptor NR5A2, which together with ESRRB is essential in maintaining pluripotency in mouse embryonic stem cells, we demonstrate conjoint bookmarking activity of both factors on promoters and enhancers of a large fraction of active genes, particularly those most efficiently reactivated in G1. Upon fast and simultaneous degradation of both factors during mitotic exit, hundreds of mitotic targets of ESRRB/NR5A2, including key players of the pluripotency network, display attenuated transcriptional reactivation. We propose that redundancy in mitotic bookmarking TFs, especially nuclear receptors, confers robustness to the reestablishment of gene regulatory networks after mitosis.


Subject(s)
Chromatin , Transcription Factors , Animals , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Mitosis/genetics , Regulatory Sequences, Nucleic Acid , Mouse Embryonic Stem Cells/metabolism
2.
Nucleic Acids Res ; 50(20): 11600-11618, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36350633

ABSTRACT

PARP1 mediates poly-ADP-ribosylation of proteins on chromatin in response to different types of DNA lesions. PARP inhibitors are used for the treatment of BRCA1/2-deficient breast, ovarian, and prostate cancer. Loss of DNA replication fork protection is proposed as one mechanism that contributes to the vulnerability of BRCA1/2-deficient cells to PARP inhibitors. However, the mechanisms that regulate PARP1 activity at stressed replication forks remain poorly understood. Here, we performed proximity proteomics of PARP1 and isolation of proteins on stressed replication forks to map putative PARP1 regulators. We identified TPX2 as a direct PARP1-binding protein that regulates the auto-ADP-ribosylation activity of PARP1. TPX2 interacts with DNA damage response proteins and promotes homology-directed repair of DNA double-strand breaks. Moreover, TPX2 mRNA levels are increased in BRCA1/2-mutated breast and prostate cancers, and high TPX2 expression levels correlate with the sensitivity of cancer cells to PARP-trapping inhibitors. We propose that TPX2 confers a mitosis-independent function in the cellular response to replication stress by interacting with PARP1.


Subject(s)
DNA Replication , Poly (ADP-Ribose) Polymerase-1 , Proteomics , DNA Breaks, Double-Stranded , DNA Repair , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
3.
Cell Mol Life Sci ; 78(14): 5587-5604, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34100981

ABSTRACT

To clarify whether differential compartmentalization of Survivin impacts temozolomide (TMZ)-triggered end points, we established a well-defined glioblastoma cell model in vitro (LN229 and A172) and in vivo, distinguishing between its nuclear and cytoplasmic localization. Expression of nuclear export sequence (NES)-mutated Survivin (SurvNESmut-GFP) led to impaired colony formation upon TMZ. This was not due to enhanced cell death but rather due to increased senescence. Nuclear-trapped Survivin reduced homologous recombination (HR)-mediated double-strand break (DSB) repair, as evaluated by γH2AX foci formation and qPCR-based HR assay leading to pronounced induction of chromosome aberrations. Opposite, clones, expressing free-shuttling cytoplasmic but not nuclear-trapped Survivin, could repair TMZ-induced DSBs and evaded senescence. Mass spectrometry-based interactomics revealed, however, no direct interaction of Survivin with any of the repair factors. The improved TMZ-triggered HR activity in Surv-GFP was associated with enhanced mRNA and stabilized RAD51 protein expression, opposite to diminished RAD51 expression in SurvNESmut cells. Notably, cytoplasmic Survivin could significantly compensate for the viability under RAD51 knockdown. Differential Survivin localization also resulted in distinctive TMZ-triggered transcriptional pathways, associated with senescence and chromosome instability as shown by global transcriptome analysis. Orthotopic LN229 xenografts, expressing SurvNESmut exhibited diminished growth and increased DNA damage upon TMZ, as manifested by PCNA and γH2AX foci expression, respectively, in brain tissue sections. Consequently, those mice lived longer. Although tumors of high-grade glioma patients expressed majorly nuclear Survivin, they exhibited rarely NES mutations which did not correlate with survival. Based on our in vitro and xenograft data, Survivin nuclear trapping would facilitate glioma response to TMZ.


Subject(s)
Cell Nucleus/metabolism , Cellular Senescence , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Homologous Recombination , Survivin/metabolism , Temozolomide/pharmacology , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis , Biomarkers, Tumor , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Nucleus/genetics , Cell Proliferation , DNA Damage , DNA Repair , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Survivin/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 116(13): 6075-6080, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30867289

ABSTRACT

Genetic mutations affecting chromatin modifiers are widespread in cancers. In malignant peripheral nerve sheath tumors (MPNSTs), Polycomb repressive complex 2 (PRC2), which plays a crucial role in gene silencing, is inactivated through recurrent mutations in core subunits embryonic ectoderm development (EED) and suppressor of zeste 12 homolog (SUZ12), but mutations in PRC2's main catalytic subunit enhancer of zeste homolog 2 (EZH2) have never been found. This is in contrast to myeloid and lymphoid malignancies, which harbor frequent loss-of-function mutations in EZH2. Here, we investigated whether the absence of EZH2 mutations in MPNST is due to a PRC2-independent (i.e., noncanonical) function of the enzyme or to redundancy with EZH1. We show that, in the absence of SUZ12, EZH2 remains bound to EED but loses its interaction with all other core and accessory PRC2 subunits. Through genetic and pharmacological analyses, we unambiguously establish that EZH2 is functionally inert in this context, thereby excluding a PRC2-independent function. Instead, we show that EZH1 and EZH2 are functionally redundant in the slowly proliferating MPNST precursors. We provide evidence that the compensatory function of EZH1 is alleviated upon higher proliferation. This work reveals how context-dependent redundancies can shape tumor-type specific mutation patterns in chromatin regulators.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Neoplasms/metabolism , Polycomb Repressive Complex 2/metabolism , Cell Line, Tumor , Cell Proliferation , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Neoplastic , Humans , Mutation/genetics , Neoplasm Proteins , Neoplasms/genetics , Neurofibroma/genetics , Neurofibroma/metabolism , Polycomb Repressive Complex 2/genetics , Transcription Factors
5.
Nat Struct Mol Biol ; 23(7): 682-690, 2016 07.
Article in English | MEDLINE | ID: mdl-27294783

ABSTRACT

Although the core subunits of Polycomb group (PcG) complexes are well characterized, little is known about the dynamics of these protein complexes during cellular differentiation. We used quantitative interaction proteomics and genome-wide profiling to study PcG proteins in mouse embryonic stem cells (ESCs) and neural progenitor cells (NPCs). We found that the stoichiometry and genome-wide binding of PRC1 and PRC2 were highly dynamic during neural differentiation. Intriguingly, we observed a downregulation and loss of PRC2 from chromatin marked with trimethylated histone H3 K27 (H3K27me3) during differentiation, whereas PRC1 was retained at these sites. Additionally, we found PRC1 at enhancer and promoter regions independently of PRC2 binding and H3K27me3. Finally, overexpression of NPC-specific PRC1 interactors in ESCs led to increased Ring1b binding to, and decreased expression of, NPC-enriched Ring1b-target genes. In summary, our integrative analyses uncovered dynamic PcG subcomplexes and their widespread colocalization with active chromatin marks during differentiation.


Subject(s)
Cell Differentiation/genetics , Chromatin/metabolism , Histones/genetics , Mouse Embryonic Stem Cells/metabolism , Neural Stem Cells/metabolism , Polycomb-Group Proteins/genetics , Animals , Cell Line , Chromatin/chemistry , Chromatography, Liquid , Chromosomes, Artificial, Bacterial , Gene Expression Regulation , Genome-Wide Association Study , Histones/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Polycomb-Group Proteins/classification , Polycomb-Group Proteins/metabolism , Protein Interaction Mapping , Proteomics/methods , Signal Transduction , Tandem Mass Spectrometry
6.
Nat Commun ; 6: 8893, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26593974

ABSTRACT

How transcription factors (TFs) cooperate within large protein complexes to allow rapid modulation of gene expression during development is still largely unknown. Here we show that the key haematopoietic LIM-domain-binding protein-1 (LDB1) TF complex contains several activator and repressor components that together maintain an erythroid-specific gene expression programme primed for rapid activation until differentiation is induced. A combination of proteomics, functional genomics and in vivo studies presented here identifies known and novel co-repressors, most notably the ETO2 and IRF2BP2 proteins, involved in maintaining this primed state. The ETO2-IRF2BP2 axis, interacting with the NCOR1/SMRT co-repressor complex, suppresses the expression of the vast majority of archetypical erythroid genes and pathways until its decommissioning at the onset of terminal erythroid differentiation. Our experiments demonstrate that multimeric regulatory complexes feature a dynamic interplay between activating and repressing components that determines lineage-specific gene expression and cellular differentiation.


Subject(s)
Carrier Proteins/metabolism , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Erythroid Cells/cytology , Erythropoiesis , Humans , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics
7.
Biochim Biophys Acta ; 1849(8): 1051-6, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26130598

ABSTRACT

A multitude of post-translational modifications take place on histones, one of the best studied being acetylation on lysine residues, which is generally associated with gene activation. During the last decades, several so-called co-repressor protein complexes that carry out the reverse process, histone deacetylation, have been identified and characterized, such as the Sin3, N-CoR/SMRT and NuRD complexes. Although a repressive role for these complexes in regulating gene expression is well established, accumulating evidence also points to a role in gene activation. Here, we argue that integration of various state-of-the-art technologies, addressing different aspects of transcriptional regulation, is essential to unravel this apparent biological versatility of 'co-repressor' complexes.


Subject(s)
Co-Repressor Proteins/physiology , Gene Expression Regulation , Multiprotein Complexes/physiology , Transcription Factors/physiology , Animals , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/physiology , Nuclear Receptor Co-Repressor 1/physiology , Nuclear Receptor Co-Repressor 2/physiology
8.
FEBS J ; 282(9): 1774-85, 2015 May.
Article in English | MEDLINE | ID: mdl-25123934

ABSTRACT

The nucleosome remodeling and deacetylase (NuRD) complex is an evolutionarily conserved chromatin-associated protein complex. Although the subunit composition of the mammalian complex is fairly well characterized, less is known about the stability and dynamics of these interactions. Furthermore, detailed information regarding protein-protein interaction surfaces within the complex is still largely lacking. Here, we show that the NuRD complex interacts with a number of substoichiometric zinc finger-containing proteins. Some of these interactions are salt-sensitive (ZNF512B and SALL4), whereas others (ZMYND8) are not. The stoichiometry of the core subunits is not affected by high salt concentrations, indicating that the core complex is stabilized by hydrophobic interactions. Interestingly, the RBBP4 and RBBP7 proteins are sensitive to high nonionic detergent concentrations during affinity purification. In a subunit exchange assay with stable isotope labeling by amino acids in cell culture (SILAC)-treated nuclear extracts, RBBP4 and RBBP7 were identified as dynamic core subunits of the NuRD complex, consistent with their proposed role as histone chaperones. Finally, using cross-linking MS, we have uncovered novel features of NuRD molecular architecture that complement our affinity purification-MS/MS data. Altogether, these findings extend our understanding of MBD3-NuRD structure and stability. STRUCTURED DIGITAL ABSTRACT: MBD3 physically interacts with ZNF512B, HDAC1, ZMYND8, GATAD2B, SALL4, GATAD2A, ZNF592, MTA3, ZNF687, CDK2AP1, CHD3, ZNF532, HDAC2, MTA2, CHD4, MTA1, KPNA2, CHD5, RBBP4 and RBBP7 by pull down (View interaction) CDK2AP1 physically interacts with MBD3, MTA3, HDAC2, GATAD2A, CHD4, CDK2AP1, MTA2, HDAC1, MTA1, CHD3, GATAD2B, MBD2, RBBP4 and RBBP7 by pull down (View interaction) MBD3 physically interacts with MTA2, MTA3, RBBP4, RBBP7, HDAC2, HDAC1, CHD4, CHD3 and MTA1 by cross-linking study (View interaction).


Subject(s)
Histone Deacetylases/metabolism , Nucleosomes/metabolism , Proteomics , Humans , Protein Binding
9.
Methods Mol Biol ; 1188: 207-26, 2014.
Article in English | MEDLINE | ID: mdl-25059614

ABSTRACT

Many cellular proteins assemble into macromolecular protein complexes. Therefore, identifying protein-protein interactions (PPIs) is essential to gain insight into the function of proteins. Recently established quantitative mass spectrometry-based techniques have significantly improved the unbiased search for PPIs. In this chapter, we describe a single-step GFP affinity purification method combined with SILAC-based quantitative mass spectrometry that can be used to identify nuclear PPIs in mammalian cells.


Subject(s)
Amino Acids/chemistry , Green Fluorescent Proteins/metabolism , Isotope Labeling/methods , Mass Spectrometry/methods , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Interaction Mapping/methods , Cell Line , Cell Nucleus/metabolism , Chromatography, Affinity , Nuclear Proteins/genetics , Nuclear Proteins/isolation & purification , Peptides/isolation & purification , Peptides/metabolism , Proteolysis , Transfection
10.
Nat Struct Mol Biol ; 21(6): 569-71, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24837194

ABSTRACT

A key step in gene repression by Polycomb is trimethylation of histone H3 K27 by PCR2 to form H3K27me3. H3K27me3 provides a binding surface for PRC1. We show that monoubiquitination of histone H2A by PRC1-type complexes to form H2Aub creates a binding site for Jarid2-Aebp2-containing PRC2 and promotes H3K27 trimethylation on H2Aub nucleosomes. Jarid2, Aebp2 and H2Aub thus constitute components of a positive feedback loop establishing H3K27me3 chromatin domains.


Subject(s)
Drosophila/genetics , Epigenetic Repression , Histones/metabolism , Polycomb-Group Proteins/genetics , Animals , Methylation , Ubiquitination
11.
Proteomics ; 14(19): 2179-89, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24634419

ABSTRACT

MBD5 and MBD6 are two members of the methyl-CpG-binding domain (MBD) family of proteins that are poorly characterized. Studies performed thus far have failed to show binding of the MBD5 and MBD6 MBD to methylated DNA. Here, we show that both MBD5 and MBD6 interact with the mammalian PR-DUB Polycomb protein complex in a mutually exclusive manner. Strikingly, the MBD of MBD5 and MBD6 is both necessary and sufficient to mediate this interaction. Chromatin immunoprecipitation analyses reveal that MBD6 and FOXK2/PR-DUB share a subset of genomic target genes, suggesting a functional interaction in vivo. Finally, we show that MBD6, but not MBD5, is recruited to sites of DNA damage in a PR-DUB independent manner. Our study thus implies a shared function for MBD5 and MBD6 through an interaction with PR-DUB, as well as an MBD6-specific recruitment to sites of DNA damage.


Subject(s)
DNA-Binding Proteins/metabolism , Polycomb-Group Proteins/metabolism , Amino Acid Sequence , Chromatin , DNA Damage , DNA Methylation , Forkhead Transcription Factors , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
12.
Methods Mol Biol ; 977: 137-57, 2013.
Article in English | MEDLINE | ID: mdl-23436359

ABSTRACT

A comprehensive identification of protein-DNA interactions that drive processes such as transcription and replication, both in prokaryotic and eukaryotic organisms, remains a major technical challenge. In this chapter, we present a SILAC-based DNA affinity purification method that can be used to identify specific interactions between proteins and functional DNA elements in an unbiased manner.


Subject(s)
DNA-Binding Proteins/isolation & purification , DNA/isolation & purification , Animals , Cell Fractionation , Cell Nucleus/chemistry , Chromatography, Affinity , DNA-Binding Proteins/chemistry , Humans , Isotope Labeling , Mass Spectrometry , Protein Binding , Proteomics , Sequence Analysis, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...