Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 171(1): 34-57, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28938122

ABSTRACT

Polycomb (PcG) and Trithorax (TrxG) group proteins are evolutionarily conserved chromatin-modifying factors originally identified as part of an epigenetic cellular memory system that maintains repressed or active gene expression states. Recently, they have been shown to globally control a plethora of cellular processes. This functional diversity is achieved by their ability to regulate chromatin at multiple levels, ranging from modifying local chromatin structure to orchestrating the three-dimensional organization of the genome. Understanding this system is a fascinating challenge of critical relevance for biology and medicine, since misexpression or mutation of multiple PcG components, as well as of TrxG members of the COMPASS family and of the SWI/SNF complex, is implicated in cancer and other diseases.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Polycomb-Group Proteins/metabolism , Animals , Chromosomal Proteins, Non-Histone/history , Embryonic Stem Cells/metabolism , Genome , History, 20th Century , History, 21st Century , Humans , Neoplasms/metabolism , Polycomb-Group Proteins/history
2.
Mol Cell ; 84(6): 1049-1061.e8, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38452766

ABSTRACT

The Polycomb repressive complex 2 (PRC2) mediates epigenetic maintenance of gene silencing in eukaryotes via methylation of histone H3 at lysine 27 (H3K27). Accessory factors define two distinct subtypes, PRC2.1 and PRC2.2, with different actions and chromatin-targeting mechanisms. The mechanisms orchestrating PRC2 assembly are not fully understood. Here, we report that alternative splicing (AS) of PRC2 core component SUZ12 generates an uncharacterized isoform SUZ12-S, which co-exists with the canonical SUZ12-L isoform in virtually all tissues and developmental stages. SUZ12-S drives PRC2.1 formation and favors PRC2 dimerization. While SUZ12-S is necessary and sufficient for the repression of target genes via promoter-proximal H3K27me3 deposition, SUZ12-L maintains global H3K27 methylation levels. Mouse embryonic stem cells (ESCs) lacking either isoform exit pluripotency more slowly and fail to acquire neuronal cell identity. Our findings reveal a physiological mechanism regulating PRC2 assembly and higher-order interactions in eutherians, with impacts on H3K27 methylation and gene repression.


Subject(s)
Alternative Splicing , Polycomb Repressive Complex 2 , Animals , Mice , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Histones/genetics , Histones/metabolism , Chromatin/genetics , Protein Isoforms/genetics
3.
Cell ; 165(5): 1224-1237, 2016 May 19.
Article in English | MEDLINE | ID: mdl-27114036

ABSTRACT

The unicellular ancestor of animals had a complex repertoire of genes linked to multicellular processes. This suggests that changes in the regulatory genome, rather than in gene innovation, were key to the origin of animals. Here, we carry out multiple functional genomic assays in Capsaspora owczarzaki, the unicellular relative of animals with the largest known gene repertoire for transcriptional regulation. We show that changing chromatin states, differential lincRNA expression, and dynamic cis-regulatory sites are associated with life cycle transitions in Capsaspora. Moreover, we demonstrate conservation of animal developmental transcription-factor networks and extensive network interconnection in this premetazoan organism. In contrast, however, Capsaspora lacks animal promoter types, and its regulatory sites are small, proximal, and lack signatures of animal enhancers. Overall, our results indicate that the emergence of animal multicellularity was linked to a major shift in genome cis-regulatory complexity, most notably the appearance of distal enhancer regulation.


Subject(s)
Biological Evolution , Eukaryota/genetics , Regulatory Elements, Transcriptional , Animals , Eukaryota/classification , Eukaryota/cytology , Gene Regulatory Networks , Genome , Histones/metabolism , Humans , Protein Processing, Post-Translational , RNA, Untranslated
4.
Mol Cell ; 83(15): 2673-2691.e7, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37506700

ABSTRACT

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


Subject(s)
Thymine DNA Glycosylase , Tumor Suppressor Protein p53 , Animals , Mice , Cell Cycle/genetics , Cell Line , Gene Expression Regulation , Thymine DNA Glycosylase/genetics , Thymine DNA Glycosylase/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Genes Dev ; 36(7-8): 451-467, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35450883

ABSTRACT

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


Subject(s)
Leukemia, Promyelocytic, Acute , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Humans , Kruppel-Like Factor 4 , Leukemia, Promyelocytic, Acute/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology
6.
Mol Cell ; 70(3): 408-421.e8, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29628311

ABSTRACT

The polycomb repressive complex 2 (PRC2) consists of core subunits SUZ12, EED, RBBP4/7, and EZH1/2 and is responsible for mono-, di-, and tri-methylation of lysine 27 on histone H3. Whereas two distinct forms exist, PRC2.1 (containing one polycomb-like protein) and PRC2.2 (containing AEBP2 and JARID2), little is known about their differential functions. Here, we report the discovery of a family of vertebrate-specific PRC2.1 proteins, "PRC2 associated LCOR isoform 1" (PALI1) and PALI2, encoded by the LCOR and LCORL gene loci, respectively. PALI1 promotes PRC2 methyltransferase activity in vitro and in vivo and is essential for mouse development. Pali1 and Aebp2 define mutually exclusive, antagonistic PRC2 subtypes that exhibit divergent H3K27-tri-methylation activities. The balance of these PRC2.1/PRC2.2 activities is required for the appropriate regulation of polycomb target genes during differentiation. PALI1/2 potentially link polycombs with transcriptional co-repressors in the regulation of cellular identity during development and in cancer.


Subject(s)
Polycomb Repressive Complex 2/genetics , Repressor Proteins/genetics , Vertebrates/genetics , Amino Acid Sequence , Animals , Cell Differentiation/genetics , Cell Line , HEK293 Cells , Histones/genetics , Humans , Methylation , Methyltransferases/genetics , Mice , Neoplasms/genetics , Sequence Alignment
7.
Mol Cell ; 65(3): 460-475.e6, 2017 Feb 02.
Article in English | MEDLINE | ID: mdl-28157506

ABSTRACT

The spatiotemporal regulation of gene expression is central for cell-lineage specification during embryonic development and is achieved through the combinatorial action of transcription factors/co-factors and epigenetic states at cis-regulatory elements. Here, we show that in addition to implementing H3K4me3 at promoters of bivalent genes, Mll2 (KMT2B)/COMPASS can also implement H3K4me3 at a subset of non-TSS regulatory elements, a subset of which shares epigenetic signatures of active enhancers. Our mechanistic studies reveal that association of Mll2's CXXC domain with CpG-rich regions plays an instrumental role for chromatin targeting and subsequent implementation of H3K4me3. Although Mll2/COMPASS is required for H3K4me3 implementation on thousands of loci, generation of catalytically mutant MLL2/COMPASS demonstrated that H3K4me3 implemented by this enzyme was essential for expression of a subset of genes, including those functioning in the control of transcriptional programs during embryonic development. Our findings suggest that not all H3K4 trimethylations implemented by MLL2/COMPASS are functionally equivalent.


Subject(s)
DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Germ Cells/cytology , Histones/metabolism , Mouse Embryonic Stem Cells/cytology , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Genetic Speciation , Germ Cells/metabolism , HEK293 Cells , Histone-Lysine N-Methyltransferase , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/chemistry , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Protein Domains
8.
Mol Cell ; 64(4): 645-658, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863225

ABSTRACT

The cellular plasticity of pluripotent stem cells is thought to be sustained by genomic regions that display both active and repressive chromatin properties. These regions exhibit low levels of gene expression, yet the mechanisms controlling these levels remain unknown. Here, we describe Elongin BC as a binding factor at the promoters of bivalent sites. Biochemical and genome-wide analyses show that Elongin BC is associated with Polycomb Repressive Complex 2 (PRC2) in pluripotent stem cells. Elongin BC is recruited to chromatin by the PRC2-associated factor EPOP (Elongin BC and Polycomb Repressive Complex 2 Associated Protein, also termed C17orf96, esPRC2p48, E130012A19Rik), a protein expressed in the inner cell mass of the mouse blastocyst. Both EPOP and Elongin BC are required to maintain low levels of expression at PRC2 genomic targets. Our results indicate that keeping the balance between activating and repressive cues is a more general feature of chromatin in pluripotent stem cells than previously appreciated.


Subject(s)
Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Pluripotent Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics , Transcription Factors/genetics , Animals , Cell Differentiation , Chromatin/chemistry , Chromatin/metabolism , Chromosomal Proteins, Non-Histone , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , Elongin , Embryo Implantation , Embryo, Mammalian , Histones/genetics , Histones/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Pluripotent Stem Cells/cytology , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , Protein Binding , Transcription Factors/metabolism , Transcription, Genetic
9.
Trends Genet ; 36(2): 118-131, 2020 02.
Article in English | MEDLINE | ID: mdl-31818514

ABSTRACT

An intricate molecular machinery is at the core of gene expression regulation in every cell. During the initial stages of organismal development, the coordinated activation of diverse transcriptional programs is crucial and must be carefully executed to shape every organ and tissue. Bivalent promoters and poised enhancers are regulatory regions decorated with histone marks that are associated with both positive and negative transcriptional outcomes. These apparently contradictory signals are important for setting bivalent genes in a poised state, which is subsequently resolved during differentiation into either active or repressive states. We discuss the origins of bivalent promoters and the mechanisms implicated in their acquisition and maintenance. We further review how the presence of bivalent marks influences genome architecture. Finally, we highlight the potential link between bivalency and cancer which could drive biomedical research in disease etiology and treatment.


Subject(s)
Cell Differentiation/genetics , Genome/genetics , Histone Code/genetics , Organogenesis/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology , Promoter Regions, Genetic/genetics
10.
Nature ; 541(7635): 41-45, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27974793

ABSTRACT

The fact that the identity of the cells that initiate metastasis in most human cancers is unknown hampers the development of antimetastatic therapies. Here we describe a subpopulation of CD44bright cells in human oral carcinomas that do not overexpress mesenchymal genes, are slow-cycling, express high levels of the fatty acid receptor CD36 and lipid metabolism genes, and are unique in their ability to initiate metastasis. Palmitic acid or a high-fat diet specifically boosts the metastatic potential of CD36+ metastasis-initiating cells in a CD36-dependent manner. The use of neutralizing antibodies to block CD36 causes almost complete inhibition of metastasis in immunodeficient or immunocompetent orthotopic mouse models of human oral cancer, with no side effects. Clinically, the presence of CD36+ metastasis-initiating cells correlates with a poor prognosis for numerous types of carcinomas, and inhibition of CD36 also impairs metastasis, at least in human melanoma- and breast cancer-derived tumours. Together, our results indicate that metastasis-initiating cells particularly rely on dietary lipids to promote metastasis.


Subject(s)
Antibodies, Neutralizing/pharmacology , CD36 Antigens/antagonists & inhibitors , Mouth Neoplasms/pathology , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , CD36 Antigens/genetics , CD36 Antigens/immunology , CD36 Antigens/metabolism , Cell Proliferation , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Lipid Metabolism/genetics , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Mice , Mouth Neoplasms/diagnosis , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Neoplastic Stem Cells/metabolism , Palmitic Acid/administration & dosage , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , Penetrance , Prognosis , Transcriptome , Xenograft Model Antitumor Assays
11.
Nature ; 544(7648): 59-64, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28289288

ABSTRACT

The folding of genomic DNA from the beads-on-a-string-like structure of nucleosomes into higher-order assemblies is crucially linked to nuclear processes. Here we calculate 3D structures of entire mammalian genomes using data from a new chromosome conformation capture procedure that allows us to first image and then process single cells. The technique enables genome folding to be examined at a scale of less than 100 kb, and chromosome structures to be validated. The structures of individual topological-associated domains and loops vary substantially from cell to cell. By contrast, A and B compartments, lamina-associated domains and active enhancers and promoters are organized in a consistent way on a genome-wide basis in every cell, suggesting that they could drive chromosome and genome folding. By studying genes regulated by pluripotency factor and nucleosome remodelling deacetylase (NuRD), we illustrate how the determination of single-cell genome structure provides a new approach for investigating biological processes.


Subject(s)
Chromatin Assembly and Disassembly , Genome , Molecular Imaging/methods , Nucleosomes/chemistry , Single-Cell Analysis/methods , Animals , CCCTC-Binding Factor , Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Mammalian/chemistry , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , Enhancer Elements, Genetic , G1 Phase , Gene Expression Regulation , Gene Regulatory Networks , Genome/genetics , Haploidy , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Models, Molecular , Molecular Conformation , Molecular Imaging/standards , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , Reproducibility of Results , Single-Cell Analysis/standards , Cohesins
12.
PLoS Comput Biol ; 17(9): e1009368, 2021 09.
Article in English | MEDLINE | ID: mdl-34473698

ABSTRACT

The ChIP-seq signal of histone modifications at promoters is a good predictor of gene expression in different cellular contexts, but whether this is also true at enhancers is not clear. To address this issue, we develop quantitative models to characterize the relationship of gene expression with histone modifications at enhancers or promoters. We use embryonic stem cells (ESCs), which contain a full spectrum of active and repressed (poised) enhancers, to train predictive models. As many poised enhancers in ESCs switch towards an active state during differentiation, predictive models can also be trained on poised enhancers throughout differentiation and in development. Remarkably, we determine that histone modifications at enhancers, as well as promoters, are predictive of gene expression in ESCs and throughout differentiation and development. Importantly, we demonstrate that their contribution to the predictive models varies depending on their location in enhancers or promoters. Moreover, we use a local regression (LOESS) to normalize sequencing data from different sources, which allows us to apply predictive models trained in a specific cellular context to a different one. We conclude that the relationship between gene expression and histone modifications at enhancers is universal and different from promoters. Our study provides new insight into how histone modifications relate to gene expression based on their location in enhancers or promoters.


Subject(s)
Enhancer Elements, Genetic , Gene Expression , Histone Code/genetics , Models, Genetic , Promoter Regions, Genetic , Animals , Cell Differentiation/genetics , Cells, Cultured , Chromatin Immunoprecipitation Sequencing/statistics & numerical data , Computational Biology , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , Regression Analysis
13.
Bioessays ; 42(12): e2000203, 2020 12.
Article in English | MEDLINE | ID: mdl-33169398

ABSTRACT

Chromatin-bound proteins underlie several fundamental cellular functions, such as control of gene expression and the faithful transmission of genetic and epigenetic information. Components of the chromatin proteome (the "chromatome") are essential in human life, and mutations in chromatin-bound proteins are frequently drivers of human diseases, such as cancer. Proteomic characterization of chromatin and de novo identification of chromatin interactors could, thus, reveal important and perhaps unexpected players implicated in human physiology and disease. Recently, intensive research efforts have focused on developing strategies to characterize the chromatome composition. In this review, we provide an overview of the dynamic composition of the chromatome, highlight the importance of its alterations as a driving force in human disease (and particularly in cancer), and discuss the different approaches to systematically characterize the chromatin-bound proteome in a global manner.


Subject(s)
Neoplasms , Proteomics , Chromatin , Epigenesis, Genetic , Humans , Neoplasms/genetics , Proteome
14.
Genes Dev ; 28(2): 182-97, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24449271

ABSTRACT

The molecular mechanisms underlying specification from embryonic stem cells (ESCs) and maintenance of neural progenitor cells (NPCs) are largely unknown. Recently, we reported that the Zuotin-related factor 1 (Zrf1) is necessary for chromatin displacement of the Polycomb-repressive complex 1 (PRC1). We found that Zrf1 is required for NPC specification from ESCs and that it promotes the expression of NPC markers, including the key regulator Pax6. Moreover, Zrf1 is essential to establish and maintain Wnt ligand expression levels, which are necessary for NPC self-renewal. Reactivation of proper Wnt signaling in Zrf1-depleted NPCs restores Pax6 expression and the self-renewal capacity. ESC-derived NPCs in vitro resemble most of the characteristics of the self-renewing NPCs located in the developing embryonic cortex, which are termed radial glial cells (RGCs). Depletion of Zrf1 in vivo impairs the expression of key self-renewal regulators and Wnt ligand genes in RGCs. Thus, we demonstrate that Zrf1 plays an essential role in NPC generation and maintenance.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Neural Plate/cytology , Neural Plate/metabolism , Oncogene Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , Ligands , Mice , Molecular Chaperones , Neurogenesis/genetics , Oncogene Proteins/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , RNA-Binding Proteins , Repressor Proteins/genetics , Signal Transduction , Wnt Proteins/metabolism
15.
Br J Cancer ; 122(3): 315-328, 2020 02.
Article in English | MEDLINE | ID: mdl-31708574

ABSTRACT

Polycomb repressive complex 2 (PRC2) is a key epigenetic multiprotein complex involved in the regulation of gene expression in metazoans. PRC2 is formed by a tetrameric core that endows the complex with histone methyltransferase activity, allowing it to mono-, di- and tri-methylate histone H3 on lysine 27 (H3K27me1/2/3); H3K27me3 is a hallmark of facultative heterochromatin. The core complex of PRC2 is bound by several associated factors that are responsible for modulating its targeting specificity and enzymatic activity. Depletion and/or mutation of the subunits of this complex can result in severe developmental defects, or even lethality. Furthermore, mutations of these proteins in somatic cells can be drivers of tumorigenesis, by altering the transcriptional regulation of key tumour suppressors or oncogenes. In this review, we present the latest results from structural studies that have characterised PRC2 composition and function. We compare this information with data and literature for both gain-of function and loss-of-function missense mutations in cancers to provide an overview of the impact of these mutations on PRC2 activity.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Neoplastic/genetics , Histone Code/genetics , Neoplasms/genetics , Polycomb Repressive Complex 2/genetics , Animals , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/ultrastructure , Gain of Function Mutation , Humans , Loss of Function Mutation , Mice , Neoplasm Proteins , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/ultrastructure , Protein Domains , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Subunits , Structure-Activity Relationship , Transcription Factors
16.
Nature ; 573(7772): 38-39, 2019 09.
Article in English | MEDLINE | ID: mdl-31481771
17.
Mol Cell ; 46(3): 369-76, 2012 05 11.
Article in English | MEDLINE | ID: mdl-22483618

ABSTRACT

Methylation of lysine 4 (K4) within histone H3 has been linked to active transcription and is removed by LSD1 and the JmjC domain-containing proteins by amino-oxidation or hydroxylation, respectively. Here, we describe the deamination catalyzed by Lysyl oxidase-like 2 protein (LOXL2) as an unconventional chemical mechanism for H3K4 modification. Infrared spectroscopy and mass spectrometry analyses demonstrated that recombinant LOXL2 specifically deaminates trimethylated H3K4. Moreover, LOXL2 activity is linked with the transcriptional control of CDH1 gene by regulating H3K4me3 deamination. These results reveal another H3 modification and provide a different mechanism for H3K4 modification.


Subject(s)
Amino Acid Oxidoreductases/physiology , Histones/metabolism , Antigens, CD , Cadherins/genetics , Cell Line, Tumor , Deamination , Gene Expression Regulation , Humans , Lysine/metabolism , Methylation
19.
Bioessays ; 38(5): 446-54, 2016 May.
Article in English | MEDLINE | ID: mdl-27000413

ABSTRACT

The Polycomb group of proteins (PcGs) are transcriptional repressor complexes that regulate important biological processes and play critical roles in cancer. Mutating or deleting EZH2 can have both oncogenic and tumor suppressive functions by increasing or decreasing H3K27me3. In contrast, mutations of SUZ12 and EED are reported to have tumor suppressive functions. EZH2 is overexpressed in many cancers, including prostate cancer, which can lead to silencing of tumor suppressors, genes regulating epithelial to mesenchymal transition (EMT), and interferon signaling. In some cases, EZH2 overexpression also leads to its use of non-histone substrates. Lastly, PRC2 associated factors can influence the progression of cancer through progressive mutations or by specific binding to certain target genes. Here, we discuss which mutations and deletions of the PRC2 complex have been detected in different cancers, with a specific focus on the overexpression of EZH2 in prostate cancer.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic , Polycomb Repressive Complex 2/genetics , Prostatic Neoplasms/genetics , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/metabolism , Epithelial-Mesenchymal Transition/genetics , Histones/genetics , Histones/metabolism , Humans , Male , Mutation , Neoplasm Proteins , Polycomb Repressive Complex 2/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction , Transcription Factors , Transcription, Genetic
20.
EMBO J ; 32(16): 2217-30, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23872946

ABSTRACT

Cellular senescence is an intrinsic defense mechanism to various cellular stresses: while still metabolically active, senescent cells stop dividing and enter a proliferation arrest. Here, we identify DPY30, a member of all mammalian histone H3K4 histone methyltransferases (HMTases), as a key regulator of the proliferation potential of human primary cells. Following depletion of DPY30, cells show a severe proliferation defect and display a senescent phenotype, including a flattened and enlarged morphology, elevated level of reactive oxygen species (ROS), increased SA-ß-galactosidase activity, and formation of senescence-associated heterochromatin foci (SAHFs). While DPY30 depletion leads to a reduced level of H3K4me3-marked active chromatin, we observed a concomitant activation of CDK inhibitors, including p16INK4a, independent of H3K4me3. ChIP experiments show that key regulators of cell-cycle progression, including ID proteins, are under direct control of DPY30. Because ID proteins are negative regulators of the transcription factors ETS1/2, depletion of DPY30 leads to the transcriptional activation of p16INK4a by ETS1/2 and thus to a senescent-like phenotype. Ectoptic re-introduction of ID protein expression can partially rescue the senescence-like phenotype induced by DPY30 depletion. Thus, our data indicate that DPY30 controls proliferation by regulating ID proteins expression, which in turn lead to senescence bypass.


Subject(s)
Cellular Senescence/physiology , Gene Expression Regulation/physiology , Inhibitor of Differentiation Protein 1/metabolism , Nuclear Proteins/metabolism , Signal Transduction/physiology , Blotting, Western , Chromatin Immunoprecipitation , Colony-Forming Units Assay , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Microarray Analysis , Nuclear Proteins/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors , beta-Galactosidase
SELECTION OF CITATIONS
SEARCH DETAIL