1301
|
Wagener N, Macher-Goeppinger S, Pritsch M, Hüsing J, Hoppe-Seyler K, Schirmacher P, Pfitzenmaier J, Haferkamp A, Hoppe-Seyler F, Hohenfellner M. Enhancer of zeste homolog 2 (EZH2) expression is an independent prognostic factor in renal cell carcinoma. BMC Cancer 2010; 10:524. [PMID: 20920340 PMCID: PMC2958940 DOI: 10.1186/1471-2407-10-524] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 10/04/2010] [Indexed: 01/08/2023] Open
Abstract
Background The enhancer of zeste homolog 2 (EZH2) gene exerts oncogene-like activities and its (over)expression has been linked to several human malignancies. Here, we studied a possible association between EZH2 expression and prognosis in patients with renal cell carcinoma (RCC). Methods EZH2 protein expression in RCC specimens was analyzed by immunohistochemistry using a tissue microarray (TMA) containing RCC tumor tissue and corresponding normal tissue samples of 520 patients. For immunohistochemical assessment of EZH2 expression, nuclear staining quantity was evaluated using a semiquantitative score. The effect of EZH2 expression on cancer specific survival (CSS) was assessed by univariate and multivariate Cox regression analyses. Results During follow-up, 147 patients (28%) had died of their disease, median follow-up of patients still alive was 6.0 years (range 0-16.1 years). EZH2 nuclear staining was present in tumor cores of 411 (79%) patients. A multivariate Cox regression analysis revealed that high nuclear EZH2 expression was an independent predictor of poor CSS (> 25-50% vs. 0%: HR 2.72, p = 0.025) in patients suffering from non-metastatic RCC. Apart from high nuclear EZH2 expression, tumor stage and Fuhrman's grading emerged as significant prognostic markers. In metastatic disease, nuclear EZH2 expression and histopathological subtype were independent predictive parameters of poor CSS (EZH2: 1-5%: HR 2.63, p = 0.043, >5-25%: HR 3.35, p = 0.013, >25%-50%: HR 4.92, p = 0.003, all compared to 0%: HR 0.36, p = 0.025, respectively). Conclusions This study defines EZH2 as a powerful independent unfavourable prognostic marker of CSS in patients with metastatic and non-metastatic RCC.
Collapse
Affiliation(s)
- Nina Wagener
- German Cancer Research Center, Molecular Therapy of Virus-Associated Cancers (F065), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1302
|
Szyf M. DNA methylation and demethylation probed by small molecules. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2010; 1799:750-9. [DOI: 10.1016/j.bbagrm.2010.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2010] [Revised: 09/05/2010] [Accepted: 09/08/2010] [Indexed: 10/19/2022]
|
1303
|
Vandegehuchte MB, De Coninck D, Vandenbrouck T, De Coen WM, Janssen CR. Gene transcription profiles, global DNA methylation and potential transgenerational epigenetic effects related to Zn exposure history in Daphnia magna. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2010; 158:3323-3329. [PMID: 20719420 DOI: 10.1016/j.envpol.2010.07.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 07/13/2010] [Accepted: 07/18/2010] [Indexed: 05/29/2023]
Abstract
A reduced level of DNA methylation has recently been described in both Zn-exposed and non-exposed offspring of Daphnia magna exposed to Zn. The hypothesis examined in this study is that DNA hypomethylation has an effect on gene transcription. A second hypothesis is that accumulative epigenetic effects can affect gene transcription in non-exposed offspring from parents with an exposure history of more than one generation. Transcriptional gene regulation was studied with a cDNA microarray. In the exposed and non-exposed hypomethylated daphnids, a large proportion of common genes were similarly up- or down-regulated, indicating a possible effect of the DNA hypomethylation. Two of these genes can be mechanistically involved in DNA methylation reduction. The similar transcriptional regulation of two and three genes in the F0 and F1 exposed daphnids on one hand and their non-exposed offspring on the other hand, could be the result of a one-generation temporary transgenerational epigenetic effect, which was not accumulative.
Collapse
Affiliation(s)
- Michiel B Vandegehuchte
- Laboratory of Environmental Toxicology and Aquatic Ecology, Ghent University, J. Plateaustraat 22, B-9000 Ghent, Belgium.
| | | | | | | | | |
Collapse
|
1304
|
Swaminathan S, Hood CL, Suzuki K, Kelleher AD. RNA duplexes in transcriptional regulation. Biomol Concepts 2010; 1:285-96. [PMID: 25962003 DOI: 10.1515/bmc.2010.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Transcriptional regulation by small RNA molecules, including small interfering RNA and microRNA, has emerged as an important gene expression modulator. The regulatory pathways controlling gene expression, post-transcriptional gene silencing and transcriptional gene silencing (TGS) have been demonstrated in yeast, plants and more recently in human cells. In this review, we discuss the currents models of transcriptional regulation and the main components of the RNA-induced silencing complex and RNA-induced transcriptional silencing complex machinery, as well as confounding off-target effects and gene activation. We also discuss RNA-mediated TGS within the NF-κB motif of the human immunodeficiency virus type 1 5' long tandem repeat promoter region and the associated epigenetic modifications. Finally, we outline the current RNA interference (RNAi) delivery methods and describe the current status of human trials investigating potential RNAi therapeutics for several human diseases.
Collapse
|
1305
|
Abstract
The discovery that cancer can be governed above and beyond the level of our DNA presents a new era for designing therapies that reverse the epigenetic state of a tumour cell. Understanding how altered chromatin dynamics leads to malignancy is essential for controlling tumour cells while sparing normal cells. Polycomb and trithorax group proteins are evolutionarily conserved and maintain chromatin in the 'off' or 'on' states, thereby preventing or promoting gene expression, respectively. Recent work highlights the dynamic interplay between these opposing classes of proteins, providing new avenues for understanding how these epigenetic regulators function in tumorigenesis.
Collapse
Affiliation(s)
- Alea A Mills
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA.
| |
Collapse
|
1306
|
Shen L, Gao G, Zhang Y, Zhang H, Ye Z, Huang S, Huang J, Kang J. A single amino acid substitution confers enhanced methylation activity of mammalian Dnmt3b on chromatin DNA. Nucleic Acids Res 2010; 38:6054-64. [PMID: 20507910 PMCID: PMC2952872 DOI: 10.1093/nar/gkq456] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2010] [Revised: 05/06/2010] [Accepted: 05/10/2010] [Indexed: 12/31/2022] Open
Abstract
Dnmt3a and Dnmt3b are paralogous enzymes responsible for de novo DNA methylation but with distinguished biological functions. In mice, disruption of Dnmt3b but not Dnmt3a causes global DNA hypomethylation, especially in repetitive sequences, which comprise the large majority of methylated DNA in the genome. By measuring DNA methylation activity of Dnmt3a and Dnmt3b homologues from five species, we found that mammalian Dnmt3b possessed significantly higher methylation activity on chromatin DNA than Dnmt3a and non-mammalian Dnmt3b. Sequence comparison and mutagenesis experiments identified a single amino acid substitution (I662N) in mammalian Dnmt3b as being crucial for its high chromatin DNA methylation activity. Further mechanistic studies demonstrated this substitution markedly enhanced the binding of Dnmt3b to nucleosomes and hence increased the chromatin DNA methylation activity. Moreover, this substitution was crucial for Dnmt3b to efficiently methylate repetitive sequences, which increased dramatically in mammalian genomes. Consistent with our observation that Dnmt3b evolved more rapidly than Dnmt3a during the emergence of mammals, these results demonstrated that the I662N substitution in mammalian Dnmt3b conferred enhanced chromatin DNA methylation activity and contributed to functional adaptation in the epigenetic system.
Collapse
Affiliation(s)
- Li Shen
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, Center for Bioinformatics, National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871 and Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Ge Gao
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, Center for Bioinformatics, National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871 and Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Ying Zhang
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, Center for Bioinformatics, National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871 and Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - He Zhang
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, Center for Bioinformatics, National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871 and Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Zhiqiang Ye
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, Center for Bioinformatics, National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871 and Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Shichao Huang
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, Center for Bioinformatics, National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871 and Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Jinyan Huang
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, Center for Bioinformatics, National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871 and Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Jiuhong Kang
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, Center for Bioinformatics, National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871 and Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| |
Collapse
|
1307
|
Choi JH, Li Y, Guo J, Pei L, Rauch TA, Kramer RS, Macmil SL, Wiley GB, Bennett LB, Schnabel JL, Taylor KH, Kim S, Xu D, Sreekumar A, Pfeifer GP, Roe BA, Caldwell CW, Bhalla KN, Shi H. Genome-wide DNA methylation maps in follicular lymphoma cells determined by methylation-enriched bisulfite sequencing. PLoS One 2010; 5:e13020. [PMID: 20927367 PMCID: PMC2947499 DOI: 10.1371/journal.pone.0013020] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Accepted: 08/21/2010] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Follicular lymphoma (FL) is a form of non-Hodgkin's lymphoma (NHL) that arises from germinal center (GC) B-cells. Despite the significant advances in immunotherapy, FL is still not curable. Beyond transcriptional profiling and genomics datasets, there currently is no epigenome-scale dataset or integrative biology approach that can adequately model this disease and therefore identify novel mechanisms and targets for successful prevention and treatment of FL. METHODOLOGY/PRINCIPAL FINDINGS We performed methylation-enriched genome-wide bisulfite sequencing of FL cells and normal CD19(+) B-cells using 454 sequencing technology. The methylated DNA fragments were enriched with methyl-binding proteins, treated with bisulfite, and sequenced using the Roche-454 GS FLX sequencer. The total number of bases covered in the human genome was 18.2 and 49.3 million including 726,003 and 1.3 million CpGs in FL and CD19(+) B-cells, respectively. 11,971 and 7,882 methylated regions of interest (MRIs) were identified respectively. The genome-wide distribution of these MRIs displayed significant differences between FL and normal B-cells. A reverse trend in the distribution of MRIs between the promoter and the gene body was observed in FL and CD19(+) B-cells. The MRIs identified in FL cells also correlated well with transcriptomic data and ChIP-on-Chip analyses of genome-wide histone modifications such as tri-methyl-H3K27, and tri-methyl-H3K4, indicating a concerted epigenetic alteration in FL cells. CONCLUSIONS/SIGNIFICANCE This study is the first to provide a large scale and comprehensive analysis of the DNA methylation sequence composition and distribution in the FL epigenome. These integrated approaches have led to the discovery of novel and frequent targets of aberrant epigenetic alterations. The genome-wide bisulfite sequencing approach developed here can be a useful tool for profiling DNA methylation in clinical samples.
Collapse
Affiliation(s)
- Jeong-Hyeon Choi
- Center of Genomics and Bioinformatics, Indiana University, Bloomington, Indiana, United States of America
| | - Yajun Li
- Medical College of Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Juyuan Guo
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Lirong Pei
- Medical College of Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Tibor A. Rauch
- Division of Biology, City of Hope Beckman Research Institute, Duarte, California, United States of America
| | - Robin S. Kramer
- Department of Computer Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Simone L. Macmil
- Advanced Center for Genome Technology, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Graham B. Wiley
- Advanced Center for Genome Technology, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Lynda B. Bennett
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Jennifer L. Schnabel
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Kristen H. Taylor
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Sun Kim
- Center of Genomics and Bioinformatics, Indiana University, Bloomington, Indiana, United States of America
| | - Dong Xu
- Division of Biology, City of Hope Beckman Research Institute, Duarte, California, United States of America
| | - Arun Sreekumar
- Medical College of Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Gerd P. Pfeifer
- Department of Computer Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Bruce A. Roe
- Advanced Center for Genome Technology, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Charles W. Caldwell
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Kapil N. Bhalla
- Medical College of Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Huidong Shi
- Medical College of Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, United States of America
| |
Collapse
|
1308
|
Shoemaker R, Wang W, Zhang K. Mediators and dynamics of DNA methylation. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2010; 3:281-98. [PMID: 20878927 DOI: 10.1002/wsbm.124] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As an inherited epigenetic marker occurring mainly on cytosines at CpG dinucleotides, DNA methylation occurs across many higher eukaryotic organisms. Looking at methylation patterns genome-wide classifies cell types uniquely and in several cases discriminates between healthy and cancerous cell types. DNA methylation can occur allele-specifically, which allows the cellular regulatory machinery to recognize each allele separately. Although only a small number of allele specifically methylated (ASM) regions are known, genome-wide experiments show that ASM is prevalent throughout the human genome. These DNA methylation patterns can be modified via DNA demethylation, which is important for induced pluripotent stem reprogramming and primordial germ cells. Recent evidence shows that the protein activation-induced cytidine deaminase plays a critical role in these demethylation events. Many transcription factors mediate DNA methylation patterns. Some transcription factors bind specifically to methylated or unmethylated sequences and other transcription factors protect genomic regions (e.g., promoter regions) from nearby DNA methylation encroachment. Possibly acting as another epigenetic regulatory layer, methylated cytosines are also converted to 5-hydroxyethylcyotines, which is a new modification type whose biological significance has yet been defined.
Collapse
Affiliation(s)
- Robert Shoemaker
- Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, CA, USA
| | | | | |
Collapse
|
1309
|
Illingworth RS, Gruenewald-Schneider U, Webb S, Kerr ARW, James KD, Turner DJ, Smith C, Harrison DJ, Andrews R, Bird AP. Orphan CpG islands identify numerous conserved promoters in the mammalian genome. PLoS Genet 2010; 6:e1001134. [PMID: 20885785 PMCID: PMC2944787 DOI: 10.1371/journal.pgen.1001134] [Citation(s) in RCA: 382] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 08/23/2010] [Indexed: 01/23/2023] Open
Abstract
CpG islands (CGIs) are vertebrate genomic landmarks that encompass the promoters of most genes and often lack DNA methylation. Querying their apparent importance, the number of CGIs is reported to vary widely in different species and many do not co-localise with annotated promoters. We set out to quantify the number of CGIs in mouse and human genomes using CXXC Affinity Purification plus deep sequencing (CAP-seq). We also asked whether CGIs not associated with annotated transcripts share properties with those at known promoters. We found that, contrary to previous estimates, CGI abundance in humans and mice is very similar and many are at conserved locations relative to genes. In each species CpG density correlates positively with the degree of H3K4 trimethylation, supporting the hypothesis that these two properties are mechanistically interdependent. Approximately half of mammalian CGIs (>10,000) are "orphans" that are not associated with annotated promoters. Many orphan CGIs show evidence of transcriptional initiation and dynamic expression during development. Unlike CGIs at known promoters, orphan CGIs are frequently subject to DNA methylation during development, and this is accompanied by loss of their active promoter features. In colorectal tumors, however, orphan CGIs are not preferentially methylated, suggesting that cancer does not recapitulate a developmental program. Human and mouse genomes have similar numbers of CGIs, over half of which are remote from known promoters. Orphan CGIs nevertheless have the characteristics of functional promoters, though they are much more likely than promoter CGIs to become methylated during development and hence lose these properties. The data indicate that orphan CGIs correspond to previously undetected promoters whose transcriptional activity may play a functional role during development.
Collapse
Affiliation(s)
- Robert S. Illingworth
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Shaun Webb
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Alastair R. W. Kerr
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Keith D. James
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Daniel J. Turner
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Colin Smith
- Neuropathology Unit, Division of Pathology, University of Edinburgh, Edinburgh, United Kingdom
| | - David J. Harrison
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Robert Andrews
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Adrian P. Bird
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
1310
|
Abstract
Emerging research suggests that long noncoding RNAs (ncRNAs) may play a role in the basic fabric of gene regulation in human cells. Mechanistic studies carried out on a small subset of antisense ncRNAs have begun to link RNA-mediated modifications of DNA and chromatin structure with gene expression, implicating ncRNAs in the regulation of transcription. Meanwhile, genome-wide studies have revealed that transcription of ncRNAs is far more ubiquitous than previously thought and suggest a more pervasive role for ncRNAs. This review will describe the current state of research regarding gene regulation by ncRNAs and highlight major techniques used in the field. Furthermore, the potential for therapeutic applications based on ncRNA regulation will also be discussed.
Collapse
|
1311
|
Cancer: evolutionary, genetic and epigenetic aspects. Clin Epigenetics 2010; 1:85-100. [PMID: 22704202 PMCID: PMC3365664 DOI: 10.1007/s13148-010-0010-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 08/31/2010] [Indexed: 12/22/2022] Open
Abstract
There exist two paradigms about the nature of cancer. According to the generally accepted one, cancer is a by-product of design limitations of a multi-cellular organism (Greaves, Nat Rev Cancer 7:213–221, 2007). The essence of the second resides in the question “Does cancer kill the individual and save the species?” (Sommer, Hum Mutat 3:166–169, 1994). Recent data on genetic and epigenetic mechanisms of cell transformation summarized in this review support the latter point of view, namely that carcinogenesis is an evolutionary conserved phenomenon—a programmed death of an organism. It is assumed that cancer possesses an important function of altruistic nature: as a mediator of negative selection, it serves to preserve integrity of species gene pool and to mediate its evolutionary adjustment. Cancer fulfills its task due apparently to specific killer function, understanding mechanism of which may suggest new therapeutic strategy.
Collapse
|
1312
|
Godmann M, May E, Kimmins S. Epigenetic mechanisms regulate stem cell expressed genes Pou5f1 and Gfra1 in a male germ cell line. PLoS One 2010; 5:e12727. [PMID: 20856864 PMCID: PMC2939054 DOI: 10.1371/journal.pone.0012727] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 08/19/2010] [Indexed: 01/15/2023] Open
Abstract
Male fertility is declining and an underlying cause may be due to environment-epigenetic interactions in developing sperm, yet nothing is known of how the epigenome controls gene expression in sperm development. Histone methylation and acetylation are dynamically regulated in spermatogenesis and are sensitive to the environment. Our objectives were to determine how histone H3 methylation and acetylation contribute to the regulation of key genes in spermatogenesis. A germ cell line, GC-1, was exposed to either the control, or the chromatin modifying drugs tranylcypromine (T), an inhibitor of the histone H3 demethylase KDM1 (lysine specific demethylase 1), or trichostatin (TSA), an inhibitor of histone deacetylases, (HDAC). Quantitative PCR (qPCR) was used to identify genes that were sensitive to treatment. As a control for specificity the Myod1 (myogenic differentiation 1) gene was analyzed. Chromatin immunoprecipitation (ChIP) followed by qPCR was used to measure histone H3 methylation and acetylation at the promoters of target genes and the control, Myod1. Remarkably, the chromatin modifying treatment specifically induced the expression of spermatogonia expressed genes Pou5f1 and Gfra1. ChIP-qPCR revealed that induction of gene expression was associated with a gain in gene activating histone H3 methylation and acetylation in Pou5f1 and Gfra1 promoters, whereas CpG DNA methylation was not affected. Our data implicate a critical role for histone H3 methylation and acetylation in the regulation of genes expressed by spermatogonia – here, predominantly mediated by HDAC-containing protein complexes.
Collapse
Affiliation(s)
- Maren Godmann
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Erin May
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Sarah Kimmins
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
1313
|
Sauvageau M, Sauvageau G. Polycomb group proteins: multi-faceted regulators of somatic stem cells and cancer. Cell Stem Cell 2010; 7:299-313. [PMID: 20804967 PMCID: PMC4959883 DOI: 10.1016/j.stem.2010.08.002] [Citation(s) in RCA: 543] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 07/30/2010] [Accepted: 08/06/2010] [Indexed: 02/06/2023]
Abstract
Polycomb Group (PcG) proteins are transcriptional repressors that epigenetically modify chromatin and participate in the establishment and maintenance of cell fates. These proteins play important roles in both stem cell self-renewal and in cancer development. Our understanding of their mechanism of action has greatly advanced over the past 10 years, but many unanswered questions remain. In this review, we present the currently available experimental data that connect PcG protein function with some of the key processes which govern somatic stem cell activity. We also highlight recent studies suggesting that a delicate balance in PcG gene dosage is crucial for proper stem cell homeostasis and prevention of cancer stem cell development.
Collapse
Affiliation(s)
- Martin Sauvageau
- Molecular Genetics of Stem Cells Laboratory, Institute of Research in Immunology and Cancer, University of Montreal, Montreal, QC H3C 3J7, Canada.
| | | |
Collapse
|
1314
|
Liu J, Zhang Z, Bando M, Itoh T, Deardorff MA, Li JR, Clark D, Kaur M, Tatsuro K, Kline AD, Chang C, Vega H, Jackson LG, Spinner NB, Shirahige K, Krantz ID. Genome-wide DNA methylation analysis in cohesin mutant human cell lines. Nucleic Acids Res 2010; 38:5657-71. [PMID: 20448023 PMCID: PMC2943628 DOI: 10.1093/nar/gkq346] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2010] [Revised: 04/19/2010] [Accepted: 04/21/2010] [Indexed: 12/17/2022] Open
Abstract
The cohesin complex has recently been shown to be a key regulator of eukaryotic gene expression, although the mechanisms by which it exerts its effects are poorly understood. We have undertaken a genome-wide analysis of DNA methylation in cohesin-deficient cell lines from probands with Cornelia de Lange syndrome (CdLS). Heterozygous mutations in NIPBL, SMC1A and SMC3 genes account for ∼65% of individuals with CdLS. SMC1A and SMC3 are subunits of the cohesin complex that controls sister chromatid cohesion, whereas NIPBL facilitates cohesin loading and unloading. We have examined the methylation status of 27 578 CpG dinucleotides in 72 CdLS and control samples. We have documented the DNA methylation pattern in human lymphoblastoid cell lines (LCLs) as well as identified specific differential DNA methylation in CdLS. Subgroups of CdLS probands and controls can be classified using selected CpG loci. The X chromosome was also found to have a unique DNA methylation pattern in CdLS. Cohesin preferentially binds to hypo-methylated DNA in control LCLs, whereas the differential DNA methylation alters cohesin binding in CdLS. Our results suggest that in addition to DNA methylation multiple mechanisms may be involved in transcriptional regulation in human cells and in the resultant gene misexpression in CdLS.
Collapse
Affiliation(s)
- Jinglan Liu
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Zhe Zhang
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Masashige Bando
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Takehiko Itoh
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Matthew A. Deardorff
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Jennifer R. Li
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Dinah Clark
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Maninder Kaur
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Kondo Tatsuro
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Antonie D. Kline
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Celia Chang
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Hugo Vega
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Laird G. Jackson
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Nancy B. Spinner
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Katsuhiko Shirahige
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Ian D. Krantz
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
1315
|
TNF inhibits Notch-1 in skeletal muscle cells by Ezh2 and DNA methylation mediated repression: implications in duchenne muscular dystrophy. PLoS One 2010; 5:e12479. [PMID: 20814569 PMCID: PMC2930001 DOI: 10.1371/journal.pone.0012479] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 08/01/2010] [Indexed: 12/21/2022] Open
Abstract
Background Classical NF-κB signaling functions as a negative regulator of skeletal myogenesis through potentially multiple mechanisms. The inhibitory actions of TNFα on skeletal muscle differentiation are mediated in part through sustained NF-κB activity. In dystrophic muscles, NF-κB activity is compartmentalized to myofibers to inhibit regeneration by limiting the number of myogenic progenitor cells. This regulation coincides with elevated levels of muscle derived TNFα that is also under IKKβ and NF-κB control. Methodology/Principal Findings Based on these findings we speculated that in DMD, TNFα secreted from myotubes inhibits regeneration by directly acting on satellite cells. Analysis of several satellite cell regulators revealed that TNFα is capable of inhibiting Notch-1 in satellite cells and C2C12 myoblasts, which was also found to be dependent on NF-κB. Notch-1 inhibition occurred at the mRNA level suggesting a transcriptional repression mechanism. Unlike its classical mode of action, TNFα stimulated the recruitment of Ezh2 and Dnmt-3b to coordinate histone and DNA methylation, respectively. Dnmt-3b recruitment was dependent on Ezh2. Conclusions/Significance We propose that in dystrophic muscles, elevated levels of TNFα and NF-κB inhibit the regenerative potential of satellite cells via epigenetic silencing of the Notch-1 gene.
Collapse
|
1316
|
Stable transmission of reversible modifications: maintenance of epigenetic information through the cell cycle. Cell Mol Life Sci 2010; 68:27-44. [PMID: 20799050 PMCID: PMC3015210 DOI: 10.1007/s00018-010-0505-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 07/19/2010] [Accepted: 08/09/2010] [Indexed: 12/30/2022]
Abstract
Even though every cell in a multicellular organism contains the same genes, the differing spatiotemporal expression of these genes determines the eventual phenotype of a cell. This means that each cell type contains a specific epigenetic program that needs to be replicated through cell divisions, along with the genome, in order to maintain cell identity. The stable inheritance of these programs throughout the cell cycle relies on several epigenetic mechanisms. In this review, DNA methylation and histone methylation by specific histone lysine methyltransferases (KMT) and the Polycomb/Trithorax proteins are considered as the primary mediators of epigenetic inheritance. In addition, non-coding RNAs and nuclear organization are implicated in the stable transfer of epigenetic information. Although most epigenetic modifications are reversible in nature, they can be stably maintained by self-recruitment of modifying protein complexes or maintenance of these complexes or structures through the cell cycle.
Collapse
|
1317
|
Abstract
The Polycomb group (PcG) of proteins is a major mechanism of epigenetic regulation that has been broadly linked to cancer. This system can repress gene expression by chromatin modification and is essential for establishing cell identity. PcG proteins are important for stem cell function and differentiation and have a profound impact during hematopoiesis. In recent years, several published studies have deepened our knowledge of the biology of the PcG in health and disease. In this article, we review the current understanding of the mechanisms of PcG-mediated repression and their relation to DNA methylation, and we discuss the role of the PcG system in hematopoiesis and hematologic malignancies. We suggest that alteration of different PcG members is a frequent event in leukemia and lymphomas that confers the stem cell properties on tumor cells. Thus, drugs targeting Polycomb complexes could be useful for treating patients with these diseases.
Collapse
|
1318
|
Abstract
Epithelial stem cells, such as those present in mammalian skin, intestine, or mammary gland, are tissue stem cells capable of both long-term self-renewal and multi-lineage differentiation. Here we review studies implicating epigenetic control mechanisms in mammalian epithelial stem cell development and homeostasis. We also provide an update of recent progresses in the involvement of canonical Wnt signaling and note an interesting link between the Wnt pathway and chromatin regulation in epithelial stem cells. We anticipate that epigenetic and epigenomic studies of these cells will increase exponentially in the near future.
Collapse
Affiliation(s)
- Bingnan Gu
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California 92697-1700
| | - Kazuhide Watanabe
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California 92697-1700
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California 92697-1700
- Developmental Biology Center, University of California, Irvine, California 92697-1700
| |
Collapse
|
1319
|
Neves R, Scheel C, Weinhold S, Honisch E, Iwaniuk KM, Trompeter HI, Niederacher D, Wernet P, Santourlidis S, Uhrberg M. Role of DNA methylation in miR-200c/141 cluster silencing in invasive breast cancer cells. BMC Res Notes 2010; 3:219. [PMID: 20682048 PMCID: PMC3161370 DOI: 10.1186/1756-0500-3-219] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 08/03/2010] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The miR-200c/141 cluster has recently been implicated in the epithelial to mesenchymal transition (EMT) process. The expression of these two miRNAs is inversely correlated with tumorigenicity and invasiveness in several human cancers. The role of these miRNAs in cancer progression is based in part on their capacity to target the EMT activators ZEB1 and ZEB2, two transcription factors, which in turn repress expression of E-cadherin. Little is known about the regulation of the mir200c/141 cluster, whose targeting has been proposed as a promising new therapy for the most aggressive tumors. FINDINGS We show that the miR-200c/141 cluster is repressed by DNA methylation of a CpG island located in the promoter region of these miRNAs. Whereas in vitro methylation of the miR-200c/141 promoter led to shutdown of promoter activity, treatment with a demethylating agent caused transcriptional reactivation in breast cancer cells formerly lacking expression of miR-200c and miR-141. More importantly, we observed that DNA methylation of the identified miR-200c/141 promoter was tightly correlated with phenotype and the invasive capacity in a panel of 8 human breast cancer cell lines. In line with this, in vitro induction of EMT by ectopic expression of the EMT transcription factor Twist in human immortalized mammary epithelial cells (HMLE) was accompanied by increased DNA methylation and concomitant repression of the miR-200c/141 locus. CONCLUSIONS The present study demonstrates that expression of the miR-200c/141 cluster is regulated by DNA methylation, suggesting epigenetic regulation of this miRNA locus in aggressive breast cancer cell lines as well as untransformed mammary epithelial cells. This epigenetic silencing mechanism might represent a novel component of the regulatory circuit for the maintenance of EMT programs in cancer and normal cells.
Collapse
Affiliation(s)
- Rui Neves
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Clinic Düsseldorf, Moorenstr, 5, Building 14,80, 40225 Düsseldorf, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1320
|
Szarc vel Szic K, Ndlovu MN, Haegeman G, Vanden Berghe W. Nature or nurture: let food be your epigenetic medicine in chronic inflammatory disorders. Biochem Pharmacol 2010; 80:1816-32. [PMID: 20688047 DOI: 10.1016/j.bcp.2010.07.029] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 07/19/2010] [Accepted: 07/23/2010] [Indexed: 01/05/2023]
Abstract
Numerous clinical, physiopathological and epidemiological studies have underlined the detrimental or beneficial role of nutritional factors in complex inflammation related disorders such as allergy, asthma, obesity, type 2 diabetes, cardiovascular disease, rheumatoid arthritis and cancer. Today, nutritional research has shifted from alleviating nutrient deficiencies to chronic disease prevention. It is known that lifestyle, environmental conditions and nutritional compounds influence gene expression. Gene expression states are set by transcriptional activators and repressors and are often locked in by cell-heritable chromatin states. Only recently, it has been observed that the environmental conditions and daily diet can affect transgenerational gene expression via "reversible" heritable epigenetic mechanisms. Epigenetic changes in DNA methylation patterns at CpG sites (epimutations) or corrupt chromatin states of key inflammatory genes and noncoding RNAs, recently emerged as major governing factors in cancer, chronic inflammatory and metabolic disorders. Reciprocally, inflammation, metabolic stress and diet composition can also change activities of the epigenetic machinery and indirectly or directly change chromatin marks. This has recently launched re-exploration of anti-inflammatory bioactive food components for characterization of their effects on epigenome modifying enzymatic activities (acetylation, methylation, phosphorylation, ribosylation, oxidation, ubiquitination, sumoylation). This may allow to improve healthy aging by reversing disease prone epimutations involved in chronic inflammatory and metabolic disorders.
Collapse
Affiliation(s)
- Katarzyna Szarc vel Szic
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Physiology, Ghent University, K.L. Ledeganckstraat 35, Gent, Belgium
| | | | | | | |
Collapse
|
1321
|
Lee CF, Ou DSC, Lee SB, Chang LH, Lin RK, Li YS, Upadhyay AK, Cheng X, Wang YC, Hsu HS, Hsiao M, Wu CW, Juan LJ. hNaa10p contributes to tumorigenesis by facilitating DNMT1-mediated tumor suppressor gene silencing. J Clin Invest 2010; 120:2920-30. [PMID: 20592467 PMCID: PMC2912195 DOI: 10.1172/jci42275] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Accepted: 05/12/2010] [Indexed: 12/25/2022] Open
Abstract
Hypermethylation-mediated tumor suppressor gene silencing plays a crucial role in tumorigenesis. Understanding its underlying mechanism is essential for cancer treatment. Previous studies on human N-alpha-acetyltransferase 10, NatA catalytic subunit (hNaa10p; also known as human arrest-defective 1 [hARD1]), have generated conflicting results with regard to its role in tumorigenesis. Here we provide multiple lines of evidence indicating that it is oncogenic. We have shown that hNaa10p overexpression correlated with poor survival of human lung cancer patients. In vitro, enforced expression of hNaa10p was sufficient to cause cellular transformation, and siRNA-mediated depletion of hNaa10p impaired cancer cell proliferation in colony assays and xenograft studies. The oncogenic potential of hNaa10p depended on its interaction with DNA methyltransferase 1 (DNMT1). Mechanistically, hNaa10p positively regulated DNMT1 enzymatic activity by facilitating its binding to DNA in vitro and its recruitment to promoters of tumor suppressor genes, such as E-cadherin, in vivo. Consistent with this, interaction between hNaa10p and DNMT1 was required for E-cadherin silencing through promoter CpG methylation, and E-cadherin repression contributed to the oncogenic effects of hNaa10p. Together, our data not only establish hNaa10p as an oncoprotein, but also reveal that it contributes to oncogenesis through modulation of DNMT1 function.
Collapse
Affiliation(s)
- Chung-Fan Lee
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Derick S.-C. Ou
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Sung-Bau Lee
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Liang-Hao Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Ruo-Kai Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Ying-Shiuan Li
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Anup K. Upadhyay
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Xiaodong Cheng
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Ching Wang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Han-Shui Hsu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Michael Hsiao
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Wen Wu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Li-Jung Juan
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
Genomics Research Center, Academia Sinica, Taipei, Taiwan.
Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.
Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
1322
|
Tsai CC, Chen CL, Liu HC, Lee YT, Wang HW, Hou LT, Hung SC. Overexpression of hTERT increases stem-like properties and decreases spontaneous differentiation in human mesenchymal stem cell lines. J Biomed Sci 2010; 17:64. [PMID: 20670406 PMCID: PMC2923118 DOI: 10.1186/1423-0127-17-64] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Accepted: 07/29/2010] [Indexed: 01/01/2023] Open
Abstract
To overcome loss of stem-like properties and spontaneous differentiation those hinder the expansion and application of human mesenchymal stem cells (hMSCs), we have clonally isolated permanent and stable human MSC lines by ectopic overexpression of primary cell cultures of hMSCs with HPV 16 E6E7 and human telomerase reverse transcriptase (hTERT) genes. These cells were found to have a differentiation potential far beyond the ordinary hMSCs. They expressed trophoectoderm and germline specific markers upon differentiation with BMP4 and retinoic acid, respectively. Furthermore, they displayed higher osteogenic and neural differentiation efficiency than primary hMSCs or hMSCs expressed HPV16 E6E7 alone with a decrease in methylation level as proven by a global CpG island methylation profile analysis. Notably, the demethylated CpG islands were highly associated with development and differentiation associated genes. Principal component analysis further pointed out the expression profile of the cells converged toward embryonic stem cells. These data demonstrate these cells not only are a useful tool for the studies of cell differentiation both for the mesenchymal and neurogenic lineages, but also provide a valuable source of cells for cell therapy studies in animal models of skeletal and neurological disorders.
Collapse
Affiliation(s)
- Chih-Chien Tsai
- Stem Cell Laboratory, Department of Medical Research & Education and Orthopaedics & Traumatology, Veterans General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
1323
|
Wu H, Coskun V, Tao J, Xie W, Ge W, Yoshikawa K, Li E, Zhang Y, Sun YE. Dnmt3a-dependent nonpromoter DNA methylation facilitates transcription of neurogenic genes. Science 2010; 329:444-8. [PMID: 20651149 PMCID: PMC3539760 DOI: 10.1126/science.1190485] [Citation(s) in RCA: 479] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
DNA methylation at proximal promoters facilitates lineage restriction by silencing cell type-specific genes. However, euchromatic DNA methylation frequently occurs in regions outside promoters. The functions of such nonproximal promoter DNA methylation are unclear. Here we show that the de novo DNA methyltransferase Dnmt3a is expressed in postnatal neural stem cells (NSCs) and is required for neurogenesis. Genome-wide analysis of postnatal NSCs indicates that Dnmt3a occupies and methylates intergenic regions and gene bodies flanking proximal promoters of a large cohort of transcriptionally permissive genes, many of which encode regulators of neurogenesis. Surprisingly, Dnmt3a-dependent nonproximal promoter methylation promotes expression of these neurogenic genes by functionally antagonizing Polycomb repression. Thus, nonpromoter DNA methylation by Dnmt3a may be used for maintaining active chromatin states of genes critical for development.
Collapse
Affiliation(s)
- Hao Wu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Volkan Coskun
- Department of Psychiatry and Biobehavioral Sciences, Intellectual Development and Disabilities Research Center at Semel Institute for Neuroscience, UCLA, Los Angeles, CA 90095, USA
| | - Jifang Tao
- Department of Psychiatry and Biobehavioral Sciences, Intellectual Development and Disabilities Research Center at Semel Institute for Neuroscience, UCLA, Los Angeles, CA 90095, USA
| | - Wei Xie
- Molecular Biology Institute, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Weihong Ge
- Department of Molecular and Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Kazuaki Yoshikawa
- Laboratory of Regulation of Neuronal Development, Institute for Protein Research, Osaka University, Suita, Osaka, 565-0871, Japan
| | - En Li
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Yi Zhang
- Howard Hughes Medical Institute, Department of Biochemistry and Biophysics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yi Eve Sun
- Department of Molecular and Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
- Department of Psychiatry and Biobehavioral Sciences, Intellectual Development and Disabilities Research Center at Semel Institute for Neuroscience, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
1324
|
Kodach LL, Jacobs RJ, Heijmans J, van Noesel CJM, Langers AMJ, Verspaget HW, Hommes DW, Offerhaus GJA, van den Brink GR, Hardwick JCH. The role of EZH2 and DNA methylation in the silencing of the tumour suppressor RUNX3 in colorectal cancer. Carcinogenesis 2010; 31:1567-75. [PMID: 20631058 PMCID: PMC2930806 DOI: 10.1093/carcin/bgq147] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In gastric cancer, a new epigenetic mechanism of tumour suppressor loss has been suggested where the histone methyltransferase enhancer of zeste homolog 2 (EZH2) is responsible for loss of expression of RUNX3. This is consistent with EZH2 upregulation in multiple cancer types being associated with poor prognosis. We investigated whether EZH2 influences the expression of RUNX3 in colorectal cancer (CRC) and whether this is independent of methylation. We determined protein and messenger RNA (mRNA) levels of EZH2 and RUNX3 and assessed RUNX3 methylation with methylation-specific polymerase chain reaction using 72 human CRCs and 8 CRC cell lines. We assessed the effect of efficient RNA interference-mediated knockdown of EZH2 on RUNX3 levels, cell viability and H3K27 trimethylation of the RUNX3 promoter using chromatin immunoprecipitation. Despite higher levels of EZH2 and lower levels of RUNX3 in CRC specimens in general, no inverse correlation between EZH2 and RUNX3 in paired samples was found arguing against a major role for histone methylation in silencing RUNX3 in CRC. Conversely, downregulation of RUNX3 mRNA in the same tumours was associated with RUNX3 DNA methylation (P < 0.05). In cell lines, knockdown of EZH2 removed the repressive chromatin marks from RUNX3 but did not result in RUNX3 re-expression. However, it prevented the re-silencing of RUNX3 after the removal of demethylating agents. In conclusion, DNA methylation is primarily responsible for the transcriptional silencing of RUNX3 in CRC, but EZH2 and histone methylation are necessary for its methylation-dependent re-silencing after the removal of demethylating agents. These results would predict that inhibitors of EZH2 and histone methylation would enhance the effects of demethylating agents in cancer therapy.
Collapse
Affiliation(s)
- Liudmila L Kodach
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1325
|
Polycomb target genes are silenced in multiple myeloma. PLoS One 2010; 5:e11483. [PMID: 20634887 PMCID: PMC2901331 DOI: 10.1371/journal.pone.0011483] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 06/02/2010] [Indexed: 11/20/2022] Open
Abstract
Multiple myeloma (MM) is a genetically heterogeneous disease, which to date remains fatal. Finding a common mechanism for initiation and progression of MM continues to be challenging. By means of integrative genomics, we identified an underexpressed gene signature in MM patient cells compared to normal counterpart plasma cells. This profile was enriched for previously defined H3K27-tri-methylated genes, targets of the Polycomb group (PcG) proteins in human embryonic fibroblasts. Additionally, the silenced gene signature was more pronounced in ISS stage III MM compared to stage I and II. Using chromatin immunoprecipitation (ChIP) assay on purified CD138+ cells from four MM patients and on two MM cell lines, we found enrichment of H3K27me3 at genes selected from the profile. As the data implied that the Polycomb-targeted gene profile would be highly relevant for pharmacological treatment of MM, we used two compounds to chemically revert the H3K27-tri-methylation mediated gene silencing. The S-adenosylhomocysteine hydrolase inhibitor 3-Deazaneplanocin (DZNep) and the histone deacetylase inhibitor LBH589 (Panobinostat), reactivated the expression of genes repressed by H3K27me3, depleted cells from the PRC2 component EZH2 and induced apoptosis in human MM cell lines. In the immunocompetent 5T33MM in vivo model for MM, treatment with LBH589 resulted in gene upregulation, reduced tumor load and increased overall survival. Taken together, our results reveal a common gene signature in MM, mediated by gene silencing via the Polycomb repressor complex. The importance of the underexpressed gene profile in MM tumor initiation and progression should be subjected to further studies.
Collapse
|
1326
|
Muthusamy V, Bosenberg M, Wajapeyee N. Redefining regulation of DNA methylation by RNA interference. Genomics 2010; 96:191-8. [PMID: 20620207 DOI: 10.1016/j.ygeno.2010.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 07/01/2010] [Accepted: 07/02/2010] [Indexed: 01/15/2023]
Abstract
Epigenetic changes refer to heritable changes that may modulate gene expression without affecting DNA sequence. DNA methylation is one such heritable epigenetic change, which is causally associated with the transcription regulation of many genes in the mammalian genome. Altered DNA methylation has been implicated in a wide variety of human diseases including cancer. Understanding the regulation of DNA methylation is likely to improve the ability to diagnose and treat these diseases. With the advent of high-throughput RNA interference (RNAi) screens, answering epigenetic questions on a genomic scale is now possible. Two recent genome-wide RNAi screens have addressed the regulation of DNA methylation in cancer, leading to the identification of the regulators of epigenetic silencing by oncogenic RAS and how epigenetic silencing of the tumor suppressor RASSF1A is maintained. These RNAi screens have much wider applications, since similar screens can now be adapted to identify the mechanism of silencing of any human disease-associated gene that is epigenetically regulated. In this review, we discuss two recent genome-wide RNAi screens for epigenetic regulators and explore potential applications in understanding DNA methylation and gene expression regulation in mammalian cells. We also discuss some of the key unanswered questions in the field of DNA methylation and suggest genome-wide RNAi screens designed to answer them.
Collapse
Affiliation(s)
- Viswanathan Muthusamy
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | |
Collapse
|
1327
|
Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet 2010; 42:722-6. [PMID: 20601953 DOI: 10.1038/ng.621] [Citation(s) in RCA: 901] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 06/03/2010] [Indexed: 12/12/2022]
Abstract
Abnormalities of chromosome 7q are common in myeloid malignancies, but no specific target genes have yet been identified. Here, we describe the finding of homozygous EZH2 mutations in 9 of 12 individuals with 7q acquired uniparental disomy. Screening of a total of 614 individuals with myeloid disorders revealed 49 monoallelic or biallelic EZH2 mutations in 42 individuals; the mutations were found most commonly in those with myelodysplastic/myeloproliferative neoplasms (27 out of 219 individuals, or 12%) and in those with myelofibrosis (4 out of 30 individuals, or 13%). EZH2 encodes the catalytic subunit of the polycomb repressive complex 2 (PRC2), a highly conserved histone H3 lysine 27 (H3K27) methyltransferase that influences stem cell renewal by epigenetic repression of genes involved in cell fate decisions. EZH2 has oncogenic activity, and its overexpression has previously been causally linked to differentiation blocks in epithelial tumors. Notably, the mutations we identified resulted in premature chain termination or direct abrogation of histone methyltransferase activity, suggesting that EZH2 acts as a tumor suppressor for myeloid malignancies.
Collapse
|
1328
|
Taylor KH, Shi H, Caldwell CW. Next generation sequencing: advances in characterizing the methylome. Genes (Basel) 2010; 1. [PMID: 24710039 PMCID: PMC3954092 DOI: 10.3390/genes1010143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Epigenetic modifications play an important role in lymphoid malignancies. This has been evidenced by the large body of work published using microarray technologies to generate methylation profiles for numerous types and subtypes of lymphoma and leukemia. These studies have shown the importance of defining the epigenome so that we can better understand the biology of lymphoma. Recent advances in DNA sequencing technology have transformed the landscape of epigenomic analysis as we now have the ability to characterize the genome-wide distribution of chromatin modifications and DNA methylation using next-generation sequencing. To take full advantage of the throughput of next-generation sequencing, there are many methodologies that have been developed and many more that are currently being developed. Choosing the appropriate methodology is fundamental to the outcome of next-generation sequencing studies. In this review, published technologies and methodologies applicable to studying the methylome are presented. In addition, progress towards defining the methylome in lymphoma is discussed and prospective directions that have been made possible as a result of next-generation sequencing technology. Finally, methodologies are introduced that have not yet been published but that are being explored in the pursuit of defining the lymphoma methylome.
Collapse
Affiliation(s)
- Kristen H Taylor
- University of Missouri-Columbia School of Medicine, Ellis Fischel Cancer Center, Columbia, MO 65212, USA.
| | - Huidong Shi
- Medical College of Georgia, Augusta, GA 30912, USA.
| | - Charles W Caldwell
- University of Missouri-Columbia School of Medicine, Ellis Fischel Cancer Center, Columbia, MO 65212, USA.
| |
Collapse
|
1329
|
Next Generation Sequencing: Advances in Characterizing the Methylome. Genes (Basel) 2010; 1:143-65. [DOI: 10.3390/genes1020143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 06/22/2010] [Accepted: 06/28/2010] [Indexed: 12/17/2022] Open
|
1330
|
Holm K, Hegardt C, Staaf J, Vallon-Christersson J, Jönsson G, Olsson H, Borg A, Ringnér M. Molecular subtypes of breast cancer are associated with characteristic DNA methylation patterns. Breast Cancer Res 2010; 12:R36. [PMID: 20565864 PMCID: PMC2917031 DOI: 10.1186/bcr2590] [Citation(s) in RCA: 221] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 05/21/2010] [Accepted: 06/18/2010] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Five different molecular subtypes of breast cancer have been identified through gene expression profiling. Each subtype has a characteristic expression pattern suggested to partly depend on cellular origin. We aimed to investigate whether the molecular subtypes also display distinct methylation profiles. METHODS We analysed methylation status of 807 cancer-related genes in 189 fresh frozen primary breast tumours and four normal breast tissue samples using an array-based methylation assay. RESULTS Unsupervised analysis revealed three groups of breast cancer with characteristic methylation patterns. The three groups were associated with the luminal A, luminal B and basal-like molecular subtypes of breast cancer, respectively, whereas cancers of the HER2-enriched and normal-like subtypes were distributed among the three groups. The methylation frequencies were significantly different between subtypes, with luminal B and basal-like tumours being most and least frequently methylated, respectively. Moreover, targets of the polycomb repressor complex in breast cancer and embryonic stem cells were more methylated in luminal B tumours than in other tumours. BRCA2-mutated tumours had a particularly high degree of methylation. Finally, by utilizing gene expression data, we observed that a large fraction of genes reported as having subtype-specific expression patterns might be regulated through methylation. CONCLUSIONS We have found that breast cancers of the basal-like, luminal A and luminal B molecular subtypes harbour specific methylation profiles. Our results suggest that methylation may play an important role in the development of breast cancers.
Collapse
Affiliation(s)
- Karolina Holm
- Department of Oncology, Clinical Sciences, Lund University, Barngatan 2B, Lund, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
1331
|
S-adenosylhomocysteine hydrolase inhibition by 3-deazaneplanocin A analogues induces anti-cancer effects in breast cancer cell lines and synergy with both histone deacetylase and HER2 inhibition. Breast Cancer Res Treat 2010; 127:109-19. [DOI: 10.1007/s10549-010-0982-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Accepted: 06/03/2010] [Indexed: 12/12/2022]
|
1332
|
Abstract
The rapid increase in incidence of obesity over the past two decades cannot be explained solely by genetic and adult lifestyle factors. There is now considerable evidence that the fetal and early postnatal environments also strongly influence the risk of developing obesity in later life. Initially, human studies showed that low birth weight was associated with an increased risk of obesity but increasingly there is evidence that overnutrition in the early life can also increase susceptibility to future obesity. These findings have now been replicated in animal models, which have shown that both maternal under- and overnutrition can induce persistent changes in gene expression and metabolism. The mechanism by which the maternal nutritional environment induces such changes is beginning to be understood and involves the altered epigenetic regulation of specific genes. In this review, we discuss the recent evidence that shows that early-life environment can induce altered epigenetic regulation leading to the induction of an altered phenotype. The demonstration of a role for altered epigenetic regulation of genes in the developmental induction of obesity opens the possibility that interventions, either through nutrition or specific drugs, may modify long-term obesity risk and combat this rapid rise in obesity.
Collapse
|
1333
|
Abstract
Genetic events alone cannot explain the entire process of carcinogenesis. It is estimated that there are more epigenetic alterations in cancer than DNA mutations, and disiphering driver and secondary events is essential to understand early processes of tumorigenesis. Epigenetic modifications control gene activity, governing whether a gene is transcribed or silent. In cancer, global patterns of two epigenetic marks, histone modifications and DNA methylation, are known to be extensively deregulated. Tumour cells are also characterized by loss-of-imprinting, a key epigenetic developmental mechanism. Genomic imprinting is the parent-of-origin, monoallelic expression of genes and is controlled by differentially DNA-methylated regions and allelic-histone modifications. With specific emphasis on imprinted loci this review will discuss alterations in DNA methylation and histone modifications in cancer. The recent advances in technology that might facilitate the identification and characterization of the epigenetic profiles of cancer will also be described.
Collapse
Affiliation(s)
- David Monk
- Imprinting and Cancer Group, Epigenetics and Cancer Biology Program, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| |
Collapse
|
1334
|
Skalska L, White RE, Franz M, Ruhmann M, Allday MJ. Epigenetic repression of p16(INK4A) by latent Epstein-Barr virus requires the interaction of EBNA3A and EBNA3C with CtBP. PLoS Pathog 2010; 6:e1000951. [PMID: 20548956 PMCID: PMC2883600 DOI: 10.1371/journal.ppat.1000951] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 05/12/2010] [Indexed: 12/14/2022] Open
Abstract
As an inhibitor of cyclin-dependent kinases, p16INK4A is an important tumour suppressor and inducer of cellular senescence that is often inactivated during the development of cancer by promoter DNA methylation. Using newly established lymphoblastoid cell lines (LCLs) expressing a conditional EBNA3C from recombinant EBV, we demonstrate that EBNA3C inactivation initiates chromatin remodelling that resets the epigenetic status of p16INK4A to permit transcriptional activation: the polycomb-associated repressive H3K27me3 histone modification is substantially reduced, while the activation-related mark H3K4me3 is modestly increased. Activation of EBNA3C reverses the distribution of these epigenetic marks, represses p16INK4A transcription and allows proliferation. LCLs lacking EBNA3A express relatively high levels of p16INK4A and have a similar pattern of histone modifications on p16INK4A as produced by the inactivation of EBNA3C. Since binding to the co-repressor of transcription CtBP has been linked to the oncogenic activity of EBNA3A and EBNA3C, we established LCLs with recombinant viruses encoding EBNA3A- and/or EBNA3C-mutants that no longer bind CtBP. These novel LCLs have revealed that the chromatin remodelling and epigenetic repression of p16INK4A requires the interaction of both EBNA3A and EBNA3C with CtBP. The repression of p16INK4A by latent EBV will not only overcome senescence in infected B cells, but may also pave the way for p16INK4A DNA methylation during B cell lymphomagenesis. We previously showed that two Epstein-Barr virus latency-associated proteins—EBNA3A and EBNA3C—contribute to enhanced B cell survival by inhibiting the expression of the death-inducing protein BIM. This repression involves remodelling of the BIM gene promoter by polycomb proteins and DNA methylation within an unusually large CpG-island that flanks the transcription initiation site. Here we show that the same two proteins, EBNA3A and EBNA3C, functionally cooperate in the polycomb-mediated chromatin remodelling of another tumour suppressor gene, p16INK4A, that encodes a cyclin-dependent kinase inhibitor capable of blocking cell proliferation. Both EBV proteins can bind the highly conserved co-repressor of transcription CtBP, and these interactions appear to be required for the efficient repression of p16INK4A. Thus by utilising the polycomb system to induce the heritable repression of two major tumour suppressor genes—one that induces cell death (BIM) and one that induces growth arrest (p16INK4A)—EBV profoundly alters latently infected B cells and their progeny, making them significantly more prone to malignant transformation.
Collapse
Affiliation(s)
- Lenka Skalska
- Section of Virology, Division of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Robert E. White
- Section of Virology, Division of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Melanie Franz
- Section of Virology, Division of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Michaela Ruhmann
- Section of Virology, Division of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Martin J. Allday
- Section of Virology, Division of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
1335
|
Günther T, Grundhoff A. The epigenetic landscape of latent Kaposi sarcoma-associated herpesvirus genomes. PLoS Pathog 2010; 6:e1000935. [PMID: 20532208 PMCID: PMC2880564 DOI: 10.1371/journal.ppat.1000935] [Citation(s) in RCA: 206] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 05/03/2010] [Indexed: 12/15/2022] Open
Abstract
Herpesvirus latency is generally thought to be governed by epigenetic modifications, but the dynamics of viral chromatin at early timepoints of latent infection are poorly understood. Here, we report a comprehensive spatial and temporal analysis of DNA methylation and histone modifications during latent infection with Kaposi Sarcoma-associated herpesvirus (KSHV), the etiologic agent of Kaposi Sarcoma and primary effusion lymphoma (PEL). By use of high resolution tiling microarrays in conjunction with immunoprecipitation of methylated DNA (MeDIP) or modified histones (chromatin IP, ChIP), our study revealed highly distinct landscapes of epigenetic modifications associated with latent KSHV infection in several tumor-derived cell lines as well as de novo infected endothelial cells. We find that KSHV genomes are subject to profound methylation at CpG dinucleotides, leading to the establishment of characteristic global DNA methylation patterns. However, such patterns evolve slowly and thus are unlikely to control early latency. In contrast, we observed that latency-specific histone modification patterns were rapidly established upon a de novo infection. Our analysis furthermore demonstrates that such patterns are not characterized by the absence of activating histone modifications, as H3K9/K14-ac and H3K4-me3 marks were prominently detected at several loci, including the promoter of the lytic cycle transactivator Rta. While these regions were furthermore largely devoid of the constitutive heterochromatin marker H3K9-me3, we observed rapid and widespread deposition of H3K27-me3 across latent KSHV genomes, a bivalent modification which is able to repress transcription in spite of the simultaneous presence of activating marks. Our findings suggest that the modification patterns identified here induce a poised state of repression during viral latency, which can be rapidly reversed once the lytic cycle is induced.
Collapse
Affiliation(s)
- Thomas Günther
- Heinrich-Pette-Institute for Experimental Virology and Immunology, Hamburg, Germany
| | - Adam Grundhoff
- Heinrich-Pette-Institute for Experimental Virology and Immunology, Hamburg, Germany
- * E-mail:
| |
Collapse
|
1336
|
Bardag-Gorce F, Li J, Oliva J, Lu SC, French BA, French SW. The cyclic pattern of blood alcohol levels during continuous ethanol feeding in rats: the effect of feeding S-adenosylmethionine. Exp Mol Pathol 2010; 88:380-7. [PMID: 20303346 PMCID: PMC2888598 DOI: 10.1016/j.yexmp.2010.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Accepted: 03/08/2010] [Indexed: 12/19/2022]
Abstract
S-adenosylmethionine (SAMe), the major methyl donor for DNA and histone methylation was fed with ethanol for 1month in order to modify the effects of ethanol on rat liver. The following parameters were studied to determine the effects of SAMe; liver histology, the blood alcohol cycle (BAL), changes in gene expression mined from microarray analysis, changes in histone methylation, changes in liver SAMe levels and its metabolites and ADH. SAMe changed the type of fatty liver, reduced liver ALT levels and prevented the BAL cycle caused by intragastric ethanol feeding. Microarray analysis showed that SAMe feeding prevented most of the changes in gene expression induced by ethanol feeding, presumably by inducing H3K27me3 and gene silencing. H3K27me3 was significantly increased by SAMe with or without ethanol feeding. It is concluded that SAMe feeding stabilized global gene expression so that the changes in gene expression involved in the blood alcohol cycle were prevented.
Collapse
Affiliation(s)
- F Bardag-Gorce
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, CA 90509, USA
| | | | | | | | | | | |
Collapse
|
1337
|
Doherty LF, Bromer JG, Zhou Y, Aldad TS, Taylor HS. In utero exposure to diethylstilbestrol (DES) or bisphenol-A (BPA) increases EZH2 expression in the mammary gland: an epigenetic mechanism linking endocrine disruptors to breast cancer. HORMONES & CANCER 2010; 1:146-55. [PMID: 21761357 PMCID: PMC3140020 DOI: 10.1007/s12672-010-0015-9] [Citation(s) in RCA: 224] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Diethylstilbestrol (DES) and bisphenol-A (BPA) are estrogen-like endocrine-disrupting chemicals that induce persistent epigenetic changes in the developing uterus. However, DES exposure in utero is also associated with an increased risk of breast cancer in adult women. Similarly, fetal exposure to BPA induces neoplastic changes in mammary tissue of mice. We hypothesized that epigenetic alterations would precede the increased risk of breast neoplasia after in utero exposure to endocrine disruptors. Enhancer of Zeste Homolog 2 (EZH2) is a histone methyltransferase that has been linked to breast cancer risk and epigenetic regulation of tumorigenesis. We examined the effect of BPA and DES on EZH2 expression and function in MCF-7 cells and in mammary glands of mice exposed in utero. DES and BPA treatment approximated human exposure. EZH2 functional activity was assessed by measuring histone H3 trimethylation. Treatment of MCF-7 cells with DES or BPA led to a 3- and 2-fold increase in EZH2 mRNA expression, respectively (p < 0.05) as well as increased EZH2 protein expression. Mice exposed to DES in utero showed a >2-fold increase in EZH2 expression in adult mammary tissue compared with controls (p < 0.05). EZH2 protein was elevated in mammary tissue of mice exposed to DES or BPA. Histone H3 trimethylation was increased in MCF-7 cells treated with BPA or DES. Similarly, mice exposed to BPA or DES in utero showed increased mammary histone H3 trimethylation. Developmental programming of EZH2 is a novel mechanism by which in utero exposure to endocrine disruptors leads to epigenetic regulation of the mammary gland.
Collapse
Affiliation(s)
- Leo F. Doherty
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, P. O. Box 208063, New Haven, CT 06520 USA
| | - Jason G. Bromer
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, P. O. Box 208063, New Haven, CT 06520 USA
| | - Yuping Zhou
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, P. O. Box 208063, New Haven, CT 06520 USA
| | - Tamir S. Aldad
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, P. O. Box 208063, New Haven, CT 06520 USA
| | - Hugh S. Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, P. O. Box 208063, New Haven, CT 06520 USA
| |
Collapse
|
1338
|
SIAH-1 interacts with mammalian polyhomeotic homologues HPH2 and affects its stability via the ubiquitin-proteasome pathway. Biochem Biophys Res Commun 2010; 397:391-6. [PMID: 20471960 DOI: 10.1016/j.bbrc.2010.05.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 05/06/2010] [Indexed: 11/23/2022]
Abstract
Polycomb Group (PcG) genes encode proteins that form large multimeric and chromatin-associated complexes implicated in the stable repression of developmentally essential genes. HPH2, the Homo sapiens polyhomeotic homologue 2, functions as one of the subunits of PcG complex 1. In our study, SIAH-1, an E3 ligase, could directly associate with HPH2 both in vitro and in vivo. Both the cysteine-rich region of SIAH-1 and the PxVxAxP motif of HPH2 were essential for the interaction. HPH2 was co-localized with SIAH-1 in nuclei. Furthermore, SIAH-1 was able to facilitate the ubiquitination and degradation of HPH2 via ubiquitin-proteasome pathway in vivo. The ubiquitination activity was severely impaired in the SIAH-1 mutant that either lost E3 ligase activity or had weakened binding ability with HPH2, strongly suggesting that SIAH-1 was the direct E3 ligase of HPH2. Thus, our results propose a novel role of SIAH-1 in regulating the expression level of HPH2 through the ubiquitin-proteasome pathway.
Collapse
|
1339
|
Identification of a PRMT5-dependent repressor complex linked to silencing of human fetal globin gene expression. Blood 2010; 116:1585-92. [PMID: 20495075 DOI: 10.1182/blood-2009-10-251116] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Defining the molecular mechanisms underpinning fetal (gamma) globin gene silencing may provide strategies for reactivation of gamma-gene expression, a major therapeutic objective in patients with beta-thalassemia and sickle cell disease (SCD). We have previously demonstrated that symmetric methylation of histone H4 Arginine 3 (H4R3me2s) by the protein arginine methyltransferase PRMT5 is required for recruitment of the DNA methyltransferase DNMT3A to the gamma-promoter, and subsequent DNA methylation and gene silencing. Here we show in an erythroid cell line, and in primary adult erythroid progenitors that PRMT5 induces additional repressive epigenetic marks at the gamma-promoter through the assembly of a multiprotein repressor complex containing the histone modifying enzymes SUV4-20h1, casein kinase 2alpha (CK2alpha), and components of the nucleosome remodeling and histone deacetylation complex. Expression of a mutant form of PRMT5 lacking methyltransferase activity or shRNA-mediated knockdown of SUV4-20h1 resulted in loss of complex binding to the gamma-promoter, reversal of both histone and DNA repressive epigenetic marks, and increased gamma-gene expression. The repressive H4K20me3 mark induced by SUV4-20h1 is enriched on the gamma-promoter in erythroid progenitors from adult bone marrow compared with cord blood, suggesting developmental specificity. These studies define coordinated epigenetic events linked to fetal globin gene silencing, and provide potential therapeutic targets for the treatment of beta-thalassemia and SCD.
Collapse
|
1340
|
Gou D, Rubalcava M, Sauer S, Mora-Bermúdez F, Erdjument-Bromage H, Tempst P, Kremmer E, Sauer F. SETDB1 is involved in postembryonic DNA methylation and gene silencing in Drosophila. PLoS One 2010; 5:e10581. [PMID: 20498723 PMCID: PMC2871795 DOI: 10.1371/journal.pone.0010581] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 03/29/2010] [Indexed: 11/19/2022] Open
Abstract
DNA methylation is fundamental for the stability and activity of genomes. Drosophila melanogaster and vertebrates establish a global DNA methylation pattern of their genome during early embryogenesis. Large-scale analyses of DNA methylation patterns have uncovered revealed that DNA methylation patterns are dynamic rather than static and change in a gene-specific fashion during development and in diseased cells. However, the factors and mechanisms involved in dynamic, postembryonic DNA methylation remain unclear. Methylation of lysine 9 in histone H3 (H3-K9) by members of the Su(var)3-9 family of histone methyltransferases (HMTs) triggers embryonic DNA methylation in Arthropods and Chordates. Here, we demonstrate that Drosophila SETDB1 (dSETDB1) can mediate DNA methylation and silencing of genes and retrotransposons. We found that dSETDB1 tri-methylates H3-K9 and binds methylated CpA motifs. Tri-methylation of H3-K9 by dSETDB1 mediates recruitment of DNA methyltransferase 2 (Dnmt2) and Su(var)205, the Drosophila ortholog of mammalian "Heterochromatin Protein 1", to target genes for dSETDB1. By enlisting Dnmt2 and Su(var)205, dSETDB1 triggers DNA methylation and silencing of genes and retrotransposons in Drosophila cells. DSETDB1 is involved in postembryonic DNA methylation and silencing of Rt1b{} retrotransposons and the tumor suppressor gene retinoblastoma family protein 1 (Rb) in imaginal discs. Collectively, our findings implicate dSETDB1 in postembryonic DNA methylation, provide a model for silencing of the tumor suppressor Rb, and uncover a role for cell type-specific DNA methylation in Drosophila development.
Collapse
Affiliation(s)
- Dawei Gou
- Department of Biochemistry, University of California Riverside, Riverside, California, United States of America
| | - Monica Rubalcava
- Department of Biochemistry, University of California Riverside, Riverside, California, United States of America
| | - Silvia Sauer
- Department of Biochemistry, University of California Riverside, Riverside, California, United States of America
| | - Felipe Mora-Bermúdez
- Zentrum für Molekulare Biologie der Universität Heidelberg, Universität Heidelberg, Heidelberg, Germany
| | - Hediye Erdjument-Bromage
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Paul Tempst
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health, München, Germany
| | - Frank Sauer
- Department of Biochemistry, University of California Riverside, Riverside, California, United States of America
- Zentrum für Molekulare Biologie der Universität Heidelberg, Universität Heidelberg, Heidelberg, Germany
| |
Collapse
|
1341
|
Hawkins RD, Hon GC, Lee LK, Ngo Q, Lister R, Pelizzola M, Edsall LE, Kuan S, Luu Y, Klugman S, Antosiewicz-Bourget J, Ye Z, Espinoza C, Agarwahl S, Shen L, Ruotti V, Wang W, Stewart R, Thomson JA, Ecker JR, Ren B. Distinct epigenomic landscapes of pluripotent and lineage-committed human cells. Cell Stem Cell 2010; 6:479-91. [PMID: 20452322 PMCID: PMC2867844 DOI: 10.1016/j.stem.2010.03.018] [Citation(s) in RCA: 628] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 01/06/2010] [Accepted: 03/12/2010] [Indexed: 01/20/2023]
Abstract
Human embryonic stem cells (hESCs) share an identical genome with lineage-committed cells, yet possess the remarkable properties of self-renewal and pluripotency. The diverse cellular properties in different cells have been attributed to their distinct epigenomes, but how much epigenomes differ remains unclear. Here, we report that epigenomic landscapes in hESCs and lineage-committed cells are drastically different. By comparing the chromatin-modification profiles and DNA methylomes in hESCs and primary fibroblasts, we find that nearly one-third of the genome differs in chromatin structure. Most changes arise from dramatic redistributions of repressive H3K9me3 and H3K27me3 marks, which form blocks that significantly expand in fibroblasts. A large number of potential regulatory sequences also exhibit a high degree of dynamics in chromatin modifications and DNA methylation. Additionally, we observe novel, context-dependent relationships between DNA methylation and chromatin modifications. Our results provide new insights into epigenetic mechanisms underlying properties of pluripotency and cell fate commitment.
Collapse
Affiliation(s)
| | - Gary C. Hon
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Leonard K. Lee
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - QueMinh Ngo
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Ryan Lister
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla CA92037, USA
| | - Mattia Pelizzola
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla CA92037, USA
| | - Lee E. Edsall
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Samantha Kuan
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Ying Luu
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Sarit Klugman
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | | | - Zhen Ye
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Celso Espinoza
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | | | - Li Shen
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Victor Ruotti
- Morgridge Institute for Research, Madison, WI 53707, USA
- Genome Center of Wisconsin, Madison, WI 53706, USA
| | - Wei Wang
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI 53707, USA
- Genome Center of Wisconsin, Madison, WI 53706, USA
| | - James A. Thomson
- Morgridge Institute for Research, Madison, WI 53707, USA
- Genome Center of Wisconsin, Madison, WI 53706, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Anatomy, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Joseph R. Ecker
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla CA92037, USA
| | - Bing Ren
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
1342
|
Zhu Q, Wani AA. Histone modifications: crucial elements for damage response and chromatin restoration. J Cell Physiol 2010; 223:283-8. [PMID: 20112283 DOI: 10.1002/jcp.22060] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Eukaryotic genomes are packaged into chromatin from repeated nucleosome arrays in which DNA sequences wrap around histones. Chromatin organization has profound influence on DNA-templated processes such as transcription, DNA replication, and repair. Recent studies have also revealed chromatin dynamics as an active contributor to diverse DNA damage responses (DDR). Here, we review recent progress in histone modification related to DDR and post-repair chromatin restoration at the sites of DNA damage. We discuss how the timing and features of histone modifications would provide the initial as well as the final guidance for DDR, and the prospect that modifications may challenge the epigenetic stability of repaired cells and serve as damage memory in chromatin.
Collapse
Affiliation(s)
- Qianzheng Zhu
- Department of Radiology, The Ohio State University, Columbus, Ohio 43240-1000, USA.
| | | |
Collapse
|
1343
|
Dambacher S, Hahn M, Schotta G. Epigenetic regulation of development by histone lysine methylation. Heredity (Edinb) 2010; 105:24-37. [PMID: 20442736 DOI: 10.1038/hdy.2010.49] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Epigenetic mechanisms contribute to the establishment and maintenance of cell-type-specific gene expression patterns. In this review, we focus on the functions of histone lysine methylation in the context of epigenetic gene regulation during developmental transitions. Over the past few years, analysis of histone lysine methylation in active and repressive nuclear compartments and, more recently, genome-wide profiling of histone lysine methylation in different cell types have revealed correlations between particular modifications and the transcriptional status of genes. Identification of histone methyltransferases (HMTases) and specific binding factors for most methylated lysine positions has provided a novel insight into the mechanisms of epigenetic gene regulation. In addition, analyses of HMTase knockout mice show that histone lysine methylation has important functions for normal development. In this study, we review mechanisms of gene activation and repression by histone lysine methylation and discuss them in the context of the developmental roles of HMTases.
Collapse
Affiliation(s)
- S Dambacher
- Munich Center for Integrated Protein Science (CiPS) and Adolf-Butenandt-Institute, Ludwig-Maximilians-University, Munich, Germany
| | | | | |
Collapse
|
1344
|
Studamire B, Goff SP. Interactions of host proteins with the murine leukemia virus integrase. Viruses 2010; 2:1110-45. [PMID: 21637732 PMCID: PMC3104679 DOI: 10.3390/v2051110] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 05/02/2010] [Accepted: 05/03/2010] [Indexed: 12/20/2022] Open
Abstract
Retroviral infections cause a variety of cancers in animals and a number of diverse diseases in humans such as leukemia and acquired immune deficiency syndrome. Productive and efficient proviral integration is critical for retroviral function and is the key step in establishing a stable and productive infection, as well as the mechanism by which host genes are activated in leukemogenesis. Host factors are widely anticipated to be involved in all stages of the retroviral life cycle, and the identification of integrase interacting factors has the potential to increase our understanding of mechanisms by which the incoming virus might appropriate cellular proteins to target and capture host DNA sequences. Identification of MoMLV integrase interacting host factors may be key to designing efficient and benign retroviral-based gene therapy vectors; key to understanding the basic mechanism of integration; and key in designing efficient integrase inhibitors. In this review, we discuss current progress in the field of MoMLV integrase interacting proteins and possible roles for these proteins in integration.
Collapse
Affiliation(s)
- Barbara Studamire
- Brooklyn College of the City University of New York, Department of Biology, 2900 Bedford Avenue, Brooklyn, NY 11210, USA; E-Mail:
| | - Stephen P. Goff
- Columbia University College of Physicians and Surgeons, Department of Biochemistry and Molecular Biophysics and Howard Hughes Medical Institute, 701 West 168 Street, New York, NY 10028, USA
| |
Collapse
|
1345
|
Karanikolas BD, Figueiredo ML, Wu L. Comprehensive evaluation of the role of EZH2 in the growth, invasion, and aggression of a panel of prostate cancer cell lines. Prostate 2010; 70:675-88. [PMID: 20087897 PMCID: PMC2848714 DOI: 10.1002/pros.21112] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Although most prostate cancers respond well to initial treatments, a fraction of prostate cancers are more aggressive and will recur and metastasize. At that point, there are few treatment options available. Significant efforts have been made to identify biomarkers that will identify these more aggressive cancers to tailor a more vigorous treatment in order to improve outcome. Polycomb Group protein enhancer of zeste 2 (EZH2) was found to be overexpressed in metastatic prostate tumors, and is considered an excellent candidate for such a biomarker. Scattered studies have found that EZH2 overexpression causes neoplastic transformation, invasion, and growth of prostate cells. However, these studies utilized different systems and cell lines, and so are difficult to correlate with one another. METHODS In this study, a comprehensive evaluation of the phenotypic effects of EZH2 in a panel of five prostate cancer cell lines was performed. By using multiple cell lines, and examining overexpression and knockdown of EZH2 concurrently, a broad view of EZH2's role in prostate cancer was achieved. RESULTS Overexpression of EZH2 led to more aggressive behaviors in all prostate cell lines tested. In contrast, downregulation of EZH2 reduced invasion and tumorigenicity of androgen-independent (AI) cell lines CWR22Rv1, PC3, and DU145, but not of androgen-dependent (AD) cell lines LAPC4 and LNCaP. CONCLUSIONS Findings from this study suggest that AI prostate tumors are more dependent on EZH2 expression than AD tumors. Our observations provide an explanation for the strong correlation between EZH2 overexpression and advanced stage, aggressive prostate cancers.
Collapse
Affiliation(s)
- Breanne D.W. Karanikolas
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Marxa L. Figueiredo
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University Baton Rouge, LA 70810 USA
| | - Lily Wu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Urology, Crump Institute for Molecular Imaging, and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
1346
|
Multiple Epigenetic Modifiers Induce Aggressive Viral Extinction in Extraembryonic Endoderm Stem Cells. Cell Stem Cell 2010; 6:457-67. [DOI: 10.1016/j.stem.2010.03.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 01/30/2010] [Accepted: 03/05/2010] [Indexed: 11/20/2022]
|
1347
|
Bredfeldt TG, Greathouse KL, Safe SH, Hung MC, Bedford MT, Walker CL. Xenoestrogen-induced regulation of EZH2 and histone methylation via estrogen receptor signaling to PI3K/AKT. Mol Endocrinol 2010; 24:993-1006. [PMID: 20351197 PMCID: PMC2870935 DOI: 10.1210/me.2009-0438] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 02/18/2010] [Indexed: 12/17/2022] Open
Abstract
Although rapid, membrane-activated estrogen receptor (ER) signaling is no longer controversial, the biological function of this nongenomic signaling is not fully characterized. We found that rapid signaling from membrane-associated ER regulates the histone methyltransferase enhancer of Zeste homolog 2 (EZH2). In response to both 17beta-estradiol (E2) and the xenoestrogen diethylstilbestrol, ER signaling via phosphatidylinositol 3-kinase/protein kinase B phosphorylates EZH2 at S21, reducing levels of trimethylation of lysine 27 on histone H3 in hormone-responsive cells. During windows of uterine development that are susceptible to developmental reprogramming, activation of this ER signaling pathway by diethylstilbestrol resulted in phosphorylation of EZH2 and reduced levels of trimethylation of lysine 27 on histone H3 in chromatin of the developing uterus. Furthermore, activation of nongenomic signaling reprogrammed the expression profile of estrogen-responsive genes in uterine myometrial cells, suggesting this as a potential mechanism for developmental reprogramming caused by early-life exposure to xenoestrogens. These data demonstrate that rapid ER signaling provides a direct linkage between xenoestrogen-induced nuclear hormone receptor signaling and modulation of the epigenetic machinery during tissue development.
Collapse
Affiliation(s)
- Tiffany G Bredfeldt
- Department of Carcinogenesis, University of Texas, M.D. Anderson Cancer Center, Science Park Research Division, 1808 Park Road 1C, P.O. Box 389, Smithville, Texas 78957, USA
| | | | | | | | | | | |
Collapse
|
1348
|
Gan Q, Schones DE, Ho Eun S, Wei G, Cui K, Zhao K, Chen X. Monovalent and unpoised status of most genes in undifferentiated cell-enriched Drosophila testis. Genome Biol 2010; 11:R42. [PMID: 20398323 PMCID: PMC2884545 DOI: 10.1186/gb-2010-11-4-r42] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Revised: 02/26/2010] [Accepted: 04/15/2010] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Increasing evidence demonstrates that stem cells maintain their identities by a unique transcription network and chromatin structure. Opposing epigenetic modifications H3K27 me3 and H3K4 me3 have been proposed to label differentiation-associated genes in stem cells, progenitor and precursor cells. In addition, many differentiation-associated genes are maintained at a poised status by recruitment of the initiative RNA Polymerase II (Pol II) at their promoter regions, in preparation for lineage-specific expression upon differentiation. Previous studies have been performed using cultured mammalian embryonic stem cells. To a lesser extent, chromatin structure has been delineated in other model organisms, such as Drosophila, to open new avenues for genetic analyses. RESULTS Here we use testes isolated from a Drosophila bag of marbles mutant strain, from which germ cells are in their undifferentiated status. We use these testes to study the endogenous chromatin structure of undifferentiated cells using ChIP-seq. We integrate the ChIP-seq with RNA-seq data, which measures the digital transcriptome. Our genome-wide analyses indicate that most differentiation-associated genes in undifferentiated cells lack an active chromatin mark and initiative Pol II; instead, they are associated with either the repressive H3K27 me3 mark or no detectable mark. CONCLUSIONS Our results reveal that most of the differentiation-associated genes in undifferentiated-cell-enriched Drosophila testes are associated with monovalent but not bivalent modifications, a chromatin signature that is distinct from the data reported in mammalian stem or precursor cells, which may reflect cell type specificity, species specificity, or both.
Collapse
Affiliation(s)
- Qiang Gan
- Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | | | | | | | | | | | | |
Collapse
|
1349
|
Siegfried Z, Simon I. DNA methylation and gene expression. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2010; 2:362-371. [DOI: 10.1002/wsbm.64] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Zahava Siegfried
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research–Israel—Canada, The Hebrew University‐Hadassah Medical School, 91120, Jerusalem, Israel
| | - Itamar Simon
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research–Israel—Canada, The Hebrew University‐Hadassah Medical School, 91120, Jerusalem, Israel
| |
Collapse
|
1350
|
Chuang LSH, Ito Y. RUNX3 is multifunctional in carcinogenesis of multiple solid tumors. Oncogene 2010; 29:2605-15. [DOI: 10.1038/onc.2010.88] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|