Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
3.
Aging Cell ; 14(6): 1113-21, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26416703

ABSTRACT

Polycomb repressive complexes (PRC1 and PRC2) are epigenetic regulators that act in coordination to influence multiple cellular processes including pluripotency, differentiation, cancer and senescence. The role of PRCs in senescence can be mostly explained by their ability to repress the INK4/ARF locus. CBX7 is one of five mammalian orthologues of Drosophila Polycomb that forms part of PRC1. Despite the relevance of CBX7 for regulating senescence and pluripotency, we have a limited understanding of how the expression of CBX7 is regulated. Here we report that the miR-9 family of microRNAs (miRNAS) downregulates the expression of CBX7. In turn, CBX7 represses miR-9-1 and miR-9-2 as part of a regulatory negative feedback loop. The miR-9/CBX7 feedback loop is a regulatory module contributing to induction of the cyclin-dependent kinase inhibitor (CDKI) p16(INK4a) during senescence. The ability of the miR-9 family to regulate senescence could have implications for understanding the role of miR-9 in cancer and aging.


Subject(s)
Aging/genetics , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation/genetics , MicroRNAs/biosynthesis , Polycomb Repressive Complex 1/biosynthesis , Cell Differentiation/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation/genetics , HEK293 Cells , Humans , MicroRNAs/genetics , Polycomb Repressive Complex 1/genetics
4.
Nucleic Acids Res ; 43(7): 3563-77, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25779048

ABSTRACT

Lytic replication of the human gamma herpes virus Epstein-Barr virus (EBV) is an essential prerequisite for the spread of the virus. Differential regulation of a limited number of cellular genes has been reported in B-cells during the viral lytic replication cycle. We asked whether a viral bZIP transcription factor, Zta (BZLF1, ZEBRA, EB1), drives some of these changes. Using genome-wide chromatin immunoprecipitation coupled to next-generation DNA sequencing (ChIP-seq) we established a map of Zta interactions across the human genome. Using sensitive transcriptome analyses we identified 2263 cellular genes whose expression is significantly changed during the EBV lytic replication cycle. Zta binds 278 of the regulated genes and the distribution of binding sites shows that Zta binds mostly to sites that are distal to transcription start sites. This differs from the prevailing view that Zta activates viral genes by binding exclusively at promoter elements. We show that a synthetic Zta binding element confers Zta regulation at a distance and that distal Zta binding sites from cellular genes can confer Zta-mediated regulation on a heterologous promoter. This leads us to propose that Zta directly reprograms the expression of cellular genes through distal elements.


Subject(s)
Gene Expression Regulation, Viral/physiology , Herpesvirus 4, Human/metabolism , Regulatory Sequences, Nucleic Acid , Trans-Activators/physiology , Base Sequence , Cell Line , Chromatin Immunoprecipitation , DNA Primers , Humans , Polymerase Chain Reaction , Transcriptome
5.
Cell Cycle ; 14(8): 1164-73, 2015.
Article in English | MEDLINE | ID: mdl-25695870

ABSTRACT

Cellular senescence, the stable cell cycle arrest elicited by various forms of stress, is an important facet of tumor suppression. Although much is known about the key players in the implementation of senescence, including the pRb and p53 axes and the cyclin dependent kinase inhibitors p16(INK4a) and p21(CIP1), many details remain unresolved. In studying conditional senescence in human fibroblasts that express a temperature sensitive SV40 large T-antigen (T-Ag), we uncovered an unexpected role for CDK4. At the permissive temperature, where pRb and p53 are functionally compromised by T-Ag, cyclin D-CDK4 complexes are disrupted by the high p16(INK4a) levels and reduced expression of p21(CIP1). In cells arrested at the non-permissive temperature, p21(CIP1) promotes reassembly of cyclin D-CDK4 yet pRb is in a hypo-phosphorylated state, consistent with cell cycle arrest. In exploring whether the reassembled cyclin D-CDK4-p21 complexes are functional, we found that shRNA-mediated knockdown or chemical inhibition of CDK4 prevented the increase in cell size associated with the senescent phenotype by allowing the cells to arrest in G1 rather than G2/M. The data point to a role for CDK4 kinase activity in a G2 checkpoint that contributes to senescence.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase 4/metabolism , Models, Biological , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Phenotype , Piperazines/pharmacology , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Retinoblastoma Protein/metabolism , Temperature , Tumor Suppressor Protein p53/metabolism
6.
Oncogene ; 34(31): 4069-4077, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-25328137

ABSTRACT

The nuclear receptor NR2E1 (also known as TLX or tailless) controls the self-renewal of neural stem cells (NSCs) and has been implied as an oncogene which initiates brain tumors including glioblastomas. Despite NR2E1 regulating targets like p21(CIP1) or PTEN we still lack a full explanation for its role in NSC self-renewal and tumorigenesis. We know that polycomb repressive complexes also control stem cell self-renewal and tumorigenesis, but so far, no formal connection has been established between NR2E1 and PRCs. In a screen for transcription factors regulating the expression of the polycomb protein CBX7, we identified NR2E1 as one of its more prominent regulators. NR2E1 binds at the CBX7 promoter, inducing its expression. Notably CBX7 represses NR2E1 as part of a regulatory loop. Ectopic NR2E1 expression inhibits cellular senescence, extending cellular lifespan in fibroblasts via CBX7-mediated regulation of p16(INK4a) and direct repression of p21(CIP1). In addition NR2E1 expression also counteracts oncogene-induced senescence. The importance of NR2E1 to restrain senescence is highlighted through the process of knocking down its expression, which causes premature senescence in human fibroblasts and epithelial cells. We also confirmed that NR2E1 regulates CBX7 and restrains senescence in NSCs. Finally, we observed that the expression of NR2E1 directly correlates with that of CBX7 in human glioblastoma multiforme. Overall we identified control of senescence and regulation of polycomb action as two possible mechanisms that can join those so far invoked to explain the role of NR2E1 in control of NSC self-renewal and cancer.


Subject(s)
Cellular Senescence/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Cells, Cultured , Feedback, Physiological , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice , Neural Stem Cells/physiology , Orphan Nuclear Receptors , Polycomb Repressive Complex 1/genetics , Promoter Regions, Genetic , Protein Binding , Receptors, Cytoplasmic and Nuclear/metabolism
7.
PLoS One ; 9(7): e102968, 2014.
Article in English | MEDLINE | ID: mdl-25057768

ABSTRACT

A growing body of evidence suggests that Polycomb group (PcG) proteins, key regulators of lineage specific gene expression, also participate in the repair of DNA double-strand breaks (DSBs) but evidence for direct recruitment of PcG proteins at specific breaks remains limited. Here we explore the association of Polycomb repressive complex 1 (PRC1) components with DSBs generated by inducible expression of the AsiSI restriction enzyme in normal human fibroblasts. Based on immunofluorescent staining, the co-localization of PRC1 proteins with components of the DNA damage response (DDR) in these primary cells is unconvincing. Moreover, using chromatin immunoprecipitation and deep sequencing (ChIP-seq), which detects PRC1 proteins at common sites throughout the genome, we did not find evidence for recruitment of PRC1 components to AsiSI-induced DSBs. In contrast, the S2056 phosphorylated form of DNA-PKcs and other DDR proteins were detected at a subset of AsiSI sites that are predominantly at the 5' ends of transcriptionally active genes. Our data question the idea that PcG protein recruitment provides a link between DSB repairs and transcriptional repression.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Activated Protein Kinase/genetics , Gene Expression Regulation , Nuclear Proteins/genetics , Polycomb Repressive Complex 1/genetics , Chromatin/chemistry , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA Restriction Enzymes/genetics , DNA Restriction Enzymes/metabolism , DNA-Activated Protein Kinase/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Histones/genetics , Histones/metabolism , Humans , Nuclear Proteins/metabolism , Phosphorylation , Polycomb Repressive Complex 1/metabolism , Primary Cell Culture , Transgenes
8.
J Cell Biol ; 204(5): 729-45, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24567357

ABSTRACT

Despite the well-documented clinical significance of the Warburg effect, it remains unclear how the aggressive glycolytic rates of tumor cells might contribute to other hallmarks of cancer, such as bypass of senescence. Here, we report that, during oncogene- or DNA damage-induced senescence, Pak1-mediated phosphorylation of phosphoglycerate mutase (PGAM) predisposes the glycolytic enzyme to ubiquitin-mediated degradation. We identify Mdm2 as a direct binding partner and ubiquitin ligase for PGAM in cultured cells and in vitro. Mutations in PGAM and Mdm2 that abrogate ubiquitination of PGAM restored the proliferative potential of primary cells under stress conditions and promoted neoplastic transformation. We propose that Mdm2, a downstream effector of p53, attenuates the Warburg effect via ubiquitination and degradation of PGAM.


Subject(s)
Cellular Senescence , Phosphoglycerate Mutase/metabolism , Proto-Oncogene Proteins c-mdm2/physiology , Stress, Physiological , Animals , Cell Line , DNA Damage , Down-Regulation , HCT116 Cells , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Phosphorylation , Proteolysis , Proto-Oncogene Proteins c-mdm2/metabolism , Ubiquitin/metabolism , p21-Activated Kinases/metabolism , p21-Activated Kinases/physiology
9.
Genome Biol ; 15(2): R23, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24485159

ABSTRACT

BACKGROUND: Polycomb group proteins form multicomponent complexes that are important for establishing lineage-specific patterns of gene expression. Mammalian cells encode multiple permutations of the prototypic Polycomb repressive complex 1 (PRC1) with little evidence for functional specialization. An aim of this study is to determine whether the multiple orthologs that are co-expressed in human fibroblasts act on different target genes and whether their genomic location changes during cellular senescence. RESULTS: Deep sequencing of chromatin immunoprecipitated with antibodies against CBX6, CBX7, CBX8, RING1 and RING2 reveals that the orthologs co-localize at multiple sites. PCR-based validation at representative loci suggests that a further six PRC1 proteins have similar binding patterns. Importantly, sequential chromatin immunoprecipitation with antibodies against different orthologs implies that multiple variants of PRC1 associate with the same DNA. At many loci, the binding profiles have a distinctive architecture that is preserved in two different types of fibroblast. Conversely, there are several hundred loci at which PRC1 binding is cell type-specific and, contrary to expectations, the presence of PRC1 does not necessarily equate with transcriptional silencing. Interestingly, the PRC1 binding profiles are preserved in senescent cells despite changes in gene expression. CONCLUSIONS: The multiple permutations of PRC1 in human fibroblasts congregate at common rather than specific sites in the genome and with overlapping but distinctive binding profiles in different fibroblasts. The data imply that the effects of PRC1 complexes on gene expression are more subtle than simply repressing the loci at which they bind.


Subject(s)
Polycomb Repressive Complex 1/biosynthesis , Polycomb-Group Proteins/biosynthesis , Protein Binding/genetics , Cell Lineage/genetics , Cellular Senescence/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental/genetics , Genome, Human , Humans , Polycomb Repressive Complex 1/genetics , Polycomb-Group Proteins/genetics
10.
J Biol Chem ; 288(51): 36398-408, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24194518

ABSTRACT

Cbx7 is one of five mammalian orthologs of the Drosophila Polycomb. Cbx7 recognizes methylated lysine residues on the histone H3 tail and contributes to gene silencing in the context of the Polycomb repressive complex 1 (PRC1). However, our knowledge of Cbx7 post-translational modifications remains limited. Through combined biochemical and mass spectrometry approaches, we report a novel phosphorylation site on mouse Cbx7 at residue Thr-118 (Cbx7T118ph), near the highly conserved Polycomb box. The generation of a site-specific antibody to Cbx7T118ph demonstrates that Cbx7 is phosphorylated via MAPK signaling. Furthermore, we find Cbx7T118 phosphorylation in murine mammary carcinoma cells, which can be blocked by MEK inhibitors. Upon EGF stimulation, Cbx7 interacts robustly with other members of PRC1. To test the role of Cbx7T118 phosphorylation in gene silencing, we employed a RAS-induced senescence model system. We demonstrate that Cbx7T118 phosphorylation moderately enhances repression of its target gene p16. In summary, we have identified and characterized a novel MAPK-mediated phosphorylation site on Cbx7 and propose that mitogen signaling to the chromatin template regulates PRC1 function.


Subject(s)
MAP Kinase Signaling System , Polycomb Repressive Complex 1/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Cellular Senescence , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Silencing , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Mutation , Phosphorylation , Polycomb Repressive Complex 1/chemistry , Polycomb Repressive Complex 1/genetics , Protein Binding , Protein Structure, Tertiary , Rats
11.
Curr Opin Cell Biol ; 25(6): 765-71, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23916530

ABSTRACT

Senescence represents a permanent exit from the cell cycle and its role in curtailing the proliferation of damaged and potentially oncogenic cells has relevance both as a front-line defense against cancer and as an underlying cause of aging. The retinoblastoma protein (RB) and p53 tumor suppressors are central to the process and the growth arrest is primarily implemented by the cyclin-dependent kinase (CDK) inhibitors, p16INK4a and p21CIP1. In contrast to terminal differentiation, senescence is a general response to a diverse range of cellular stresses and is typically accompanied by a characteristic set of phenotypic changes. Of particular note is a secretory program whose autocrine and paracrine effects can advertize the presence of senescent cells within a tissue and promote their clearance by the immune system. In this short review, we will highlight recent advances in understanding the relationship between senescence and aging and the distinction between senescence and terminal differentiation, from a cell cycle perspective.


Subject(s)
Aging , Cell Cycle , Cellular Senescence , Aging/genetics , Animals , Cell Cycle/genetics , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage , DNA Replication/genetics , Down-Regulation/genetics , E2F Transcription Factors/metabolism , Epigenesis, Genetic/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
12.
EMBO J ; 32(7): 982-95, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23455154

ABSTRACT

The INK4/ARF locus regulates senescence and is frequently altered in cancer. In normal cells, the INK4/ARF locus is found silenced by Polycomb repressive complexes (PRCs). Which are the mechanisms responsible for the recruitment of PRCs to INK4/ARF and their other target genes remains unclear. In a genetic screen for transcription factors regulating senescence, we identified the homeodomain-containing protein HLX1 (H2.0-like homeobox 1). Expression of HLX1 extends cellular lifespan and blunts oncogene-induced senescence. Using quantitative proteomics, we identified p16(INK4a) as the key target mediating the effects of HLX1 in senescence. HLX1 represses p16(INK4a) transcription by recruiting PRCs and HDAC1. This mechanism has broader implications, as HLX1 also regulates a subset of PRC targets besides p16(INK4a). Finally, sampling members of the Homeobox family, we identified multiple genes with ability to repress p16(INK4a). Among them, we found HOXA9 (Homeobox A9), a putative oncogene in leukaemia, which also recruits PRCs and HDAC1 to regulate p16(INK4a). Our results reveal an unexpected and conserved interplay between homeodomain-containing proteins and PRCs with implications in senescence, development and cancer.


Subject(s)
Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Polycomb-Group Proteins/metabolism , Transcription Factors/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , HeLa Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Homeodomain Proteins/genetics , Humans , Polycomb-Group Proteins/genetics , Transcription Factors/genetics
13.
Cell Stem Cell ; 10(1): 33-46, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22226354

ABSTRACT

The Polycomb Group (PcG) of chromatin modifiers regulates pluripotency and differentiation. Mammalian genomes encode multiple homologs of the Polycomb repressive complex 1 (PRC1) components, including five orthologs of the Drosophila Polycomb protein (Cbx2, Cbx4, Cbx6, Cbx7, and Cbx8). We have identified Cbx7 as the primary Polycomb ortholog of PRC1 complexes in embryonic stem cells (ESCs). The expression of Cbx7 is downregulated during ESC differentiation, preceding the upregulation of Cbx2, Cbx4, and Cbx8, which are directly repressed by Cbx7. Ectopic expression of Cbx7 inhibits differentiation and X chromosome inactivation and enhances ESC self-renewal. Conversely, Cbx7 knockdown induces differentiation and derepresses lineage-specific markers. In a functional screen, we identified the miR-125 and miR-181 families as regulators of Cbx7 that are induced during ESC differentiation. Ectopic expression of these miRNAs accelerates ESC differentiation via regulation of Cbx7. These observations establish a critical role for Cbx7 and its regulatory miRNAs in determining pluripotency.


Subject(s)
Cell Differentiation/physiology , Down-Regulation/physiology , Embryonic Stem Cells/metabolism , MicroRNAs/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/metabolism , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Cell Line, Tumor , Chromosomes, Human, X/genetics , Chromosomes, Human, X/metabolism , Embryonic Stem Cells/cytology , Humans , Ligases , Mice , MicroRNAs/genetics , Mitochondrial Membrane Transport Proteins , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Repressor Proteins/genetics , Ubiquitin-Protein Ligases , X Chromosome Inactivation/physiology
14.
Mol Cell ; 45(1): 123-31, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22178396

ABSTRACT

Both the DNA damage response (DDR) and epigenetic mechanisms play key roles in the implementation of senescent phenotypes, but very little is known about how these two mechanisms are integrated to establish senescence-associated gene expression. Here we show that, in senescent cells, the DDR induces proteasomal degradation of G9a and GLP, major histone H3K9 mono- and dimethyltransferases, through Cdc14B- and p21(Waf1/Cip1)-dependent activation of APC/C(Cdh1) ubiquitin ligase, thereby causing a global decrease in H3K9 dimethylation, an epigenetic mark for euchromatic gene silencing. Interestingly, induction of IL-6 and IL-8, major players of the senescence-associated secretory phenotype (SASP), correlated with a decline of H3K9 dimethylation around the respective gene promoters and knockdown of Cdh1 abolished IL-6/IL-8 expression in senescent cells, suggesting that the APC/C(Cdh1)-G9a/GLP axis plays crucial roles in aspects of senescent phenotype. These findings establish a role for APC/C(Cdh1) and reveal how the DDR integrates with epigenetic processes to induce senescence-associated gene expression.


Subject(s)
Cellular Senescence , DNA Damage , Histone-Lysine N-Methyltransferase/metabolism , Ubiquitin-Protein Ligase Complexes/physiology , Anaphase-Promoting Complex-Cyclosome , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Dual-Specificity Phosphatases/physiology , Histocompatibility Antigens/metabolism , Histone Methyltransferases , Histones/metabolism , Humans , Methylation , Signal Transduction
15.
Cancer Res ; 72(1): 165-75, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22080569

ABSTRACT

The cyclin-dependent kinase (CDK) inhibitors, p18(INK4c) and p16(INK4a), both have the credentials of tumor suppressors in human cancers and mouse models. For p16(INK4a), the underlying rationale is its role in senescence, but the selective force for inactivation of p18(INK4c) in incipient cancer cells is less clear. Here, we show that in human fibroblasts undergoing replicative or oncogene-induced senescence, there is a marked decline in the levels of p18(INK4c) protein and RNA, which mirrors the accumulation of p16(INK4a). Downregulation of INK4c is not dependent on p16(INK4a), and RAS can promote the loss of INK4c without cell-cycle arrest. Downregulation of p18(INK4c) correlates with reduced expression of menin and E2F1 but is unaffected by acute cell-cycle arrest or inactivation of the retinoblastoma protein (pRb). Collectively, our data question the idea that p18(INK4c) acts as a backup for loss of p16(INK4a) and suggest that the apparent activation of p18(INK4c) in some settings represents delayed senescence rather than increased expression. We propose that the contrasting behavior of the two very similar INK4 proteins could reflect their respective roles in senescence versus differentiation.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/physiology , Cyclin-Dependent Kinase Inhibitor p18/physiology , Genes, Tumor Suppressor , Oncogenes , Base Sequence , Down-Regulation , Humans , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
16.
EMBO J ; 29(15): 2553-65, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20601937

ABSTRACT

An important facet of transcriptional repression by Polycomb repressive complex 1 (PRC1) is the mono-ubiquitination of histone H2A by the combined action of the Posterior sex combs (Psc) and Sex combs extra (Sce) proteins. Here, we report that two ubiquitin-specific proteases, USP7 and USP11, co-purify with human PRC1-type complexes through direct interactions with the Psc orthologues MEL18 and BMI1, and with other PRC1 components. Ablation of either USP7 or USP11 in primary human fibroblasts results in de-repression of the INK4a tumour suppressor accompanied by loss of PRC1 binding at the locus and a senescence-like proliferative arrest. Mechanistically, USP7 and USP11 regulate the ubiquitination status of the Psc and Sce proteins themselves, thereby affecting their turnover and abundance. Our results point to a novel function for USPs in the regulation and function of Polycomb complexes.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Repressor Proteins/metabolism , Thiolester Hydrolases/metabolism , Ubiquitin Thiolesterase/metabolism , Cell Proliferation , Cells, Cultured , Histones/metabolism , Humans , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Protein Binding , Proto-Oncogene Proteins/metabolism , RNA Interference , Thiolester Hydrolases/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitination
17.
Nat Struct Mol Biol ; 17(7): 862-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20543829

ABSTRACT

Several lines of evidence point to a role for noncoding RNA in transcriptional repression by Polycomb group (PcG) proteins, but the precise mechanism remains unclear. Here we show that human MOV10, a putative RNA helicase previously implicated in post-transcriptional gene silencing, co-purifies and interacts with components of Polycomb-repressive complex 1 (PRC1) from human cells. Endogenous human MOV10 is mostly nuclear, and a proportion associates with chromatin in an RNA-dependent manner. Small hairpin RNA (shRNA)-mediated knockdown of MOV10 in human fibroblasts leads to the upregulation of the INK4a tumor suppressor, a known target of PcG-mediated repression, accompanied by the dissociation of PRC1 proteins from the locus and a reduction in trimethylation of histone H3 on Lys27 (H3K27me3). As well as prompting reassessment of MOV10's role in other settings, our findings suggest that it is directly involved in transcriptional silencing by PcG complexes.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Silencing , RNA Helicases/metabolism , Repressor Proteins/metabolism , Cell Line , Cells, Cultured , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Fibroblasts/metabolism , Gene Knockdown Techniques , Humans , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , RNA Helicases/genetics
18.
PLoS One ; 4(7): e6380, 2009 Jul 28.
Article in English | MEDLINE | ID: mdl-19636380

ABSTRACT

Misexpression of Polycomb repressive complex 1 (PRC1) components in human cells profoundly influences the onset of cellular senescence by modulating transcription of the INK4a tumor suppressor gene. Using tandem affinity purification, we find that CBX7 and CBX8, two Polycomb (Pc) homologs that repress INK4a, both participate in PRC1-like complexes with at least two Posterior sex combs (Psc) proteins, MEL18 and BMI1. Each complex contains a single representative of the Pc and Psc families. In primary human fibroblasts, CBX7, CBX8, MEL18 and BMI1 are present at the INK4a locus and shRNA-mediated knockdown of any one of these components results in de-repression of INK4a and proliferative arrest. Sequential chromatin immunoprecipitation (ChIP) reveals that CBX7 and CBX8 bind simultaneously to the same region of chromatin and knockdown of one of the Pc or Psc proteins results in release of the other, suggesting that the binding of PRC1 complexes is interdependent. Our findings provide the first evidence that a single gene can be regulated by several distinct PRC1 complexes and raise important questions about their configuration and relative functions.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Genes, Tumor Suppressor , Cells, Cultured , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Protein Binding
19.
Genes Dev ; 23(10): 1177-82, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19451218

ABSTRACT

The INK4a/ARF tumor suppressor locus, a key executor of cellular senescence, is regulated by members of the Polycomb group (PcG) of transcriptional repressors. Here we show that signaling from oncogenic RAS overrides PcG-mediated repression of INK4a by activating the H3K27 demethylase JMJD3 and down-regulating the methyltransferase EZH2. In human fibroblasts, JMJD3 activates INK4a, but not ARF, and causes p16(INK4a)-dependent arrest. In mouse embryo fibroblasts, Jmjd3 activates both Ink4a and Arf and elicits a p53-dependent arrest, echoing the effects of RAS in this system. Our findings directly implicate JMJD3 in the regulation of INK4a/ARF during oncogene-induced senescence and suggest that JMJD3 has the capacity to act as a tumor suppressor.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Epigenesis, Genetic/genetics , Gene Expression Regulation , Oxidoreductases, N-Demethylating/metabolism , ras Proteins/metabolism , Animals , Cellular Senescence , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases , Mice , Signal Transduction
20.
PLoS One ; 4(4): e5067, 2009.
Article in English | MEDLINE | ID: mdl-19340300

ABSTRACT

The CDKN2A locus encodes two important tumor suppressors, INK4a and ARF, which respond to oncogenic stresses by inducing cellular senescence. We conducted a genome-scale cDNA overexpression screen using a reporter containing INK4a regulatory sequences to identify novel transcriptional activators of this locus. This screen revealed 285 cDNAs that putatively regulate the transcriptional activation of INK4a. Of these, 56 are annotated as transcription factors, including two previously reported activators of the locus, ETS2 and JUNB. Fourteen genes were further validated for activity and specificity, including several homeodomain proteins. We found that the transcription of one of these, the homeodomain protein MEOX2 (GAX) is enhanced in primary cells during the induction of senescence, and forced expression of this protein results in the induction of premature senescence. We further demonstrate that MEOX2-induced senescence is dependent upon INK4a activity, and chromatin immunoprecipitation studies indicate that MEOX2 directly binds the INK4a promoter. These results support a role for this homeodomain protein as a direct regulator of INK4a transcription and senescence in human cells.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/physiology , Homeodomain Proteins/physiology , Trans-Activators/physiology , Base Sequence , Cells, Cultured , DNA Primers , DNA, Complementary , Humans , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...