69
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      MBD3 Localizes at Promoters, Gene Bodies and Enhancers of Active Genes

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The Mi-2/nucleosome remodeling and histone deacetylase (NuRD) complex is a multiprotein machine proposed to regulate chromatin structure by nucleosome remodeling and histone deacetylation activities. Recent reports describing localization of NuRD provide new insights that question previous models on NuRD action, but are not in complete agreement. Here, we provide location analysis of endogenous MBD3, a component of NuRD complex, in two human breast cancer cell lines (MCF-7 and MDA-MB-231) using two independent genomic techniques: DNA adenine methyltransferase identification (DamID) and ChIP-seq. We observed concordance of the resulting genomic localization, suggesting that these studies are converging on a robust map for NuRD in the cancer cell genome. MBD3 preferentially associated with CpG rich promoters marked by H3K4me3 and showed cell-type specific localization across gene bodies, peaking around the transcription start site. A subset of sites bound by MBD3 was enriched in H3K27ac and was in physical proximity to promoters in three-dimensional space, suggesting function as enhancers. MBD3 enrichment was also noted at promoters modified by H3K27me3. Functional analysis of chromatin indicated that MBD3 regulates nucleosome occupancy near promoters and in gene bodies. These data suggest that MBD3, and by extension the NuRD complex, may have multiple roles in fine tuning expression for both active and silent genes, representing an important step in defining regulatory mechanisms by which NuRD complex controls chromatin structure and modification status.

          Author Summary

          Chromatin structure is tightly regulated by multiple mechanisms; its dysregulation is associated with developmental abnormalities and disease. The Mi-2/nucleosome remodeling and histone deacetylase (NuRD) complex is proposed to regulate chromatin structure by changing the location and/or the chemical properties of the fundamental building block of chromatin, the nucleosome. NuRD has been shown by genetics to be important for normal development, yet the detailed mechanism of how NuRD regulates chromatin structure is still unclear. Here, we study the localization and function of MBD3, a component of NuRD, in two human breast cancer cell lines using two independent genomic technologies. Our data demonstrate that existing models, which associate NuRD with transcriptional repression, are not completely correct. Rather, MBD3 showed cell-type specific localization at active genes. Moreover, we found a previously unidentified localization of MBD3 across gene bodies and identified a regulatory role for MBD3 in nucleosome organization. Our data provide a reliable starting point from which to address mechanisms by which NuRD controls chromatin structure and nuclear biology.

          Related collections

          Most cited references35

          • Record: found
          • Abstract: found
          • Article: not found

          A clustering approach for identification of enriched domains from histone modification ChIP-Seq data.

          Chromatin states are the key to gene regulation and cell identity. Chromatin immunoprecipitation (ChIP) coupled with high-throughput sequencing (ChIP-Seq) is increasingly being used to map epigenetic states across genomes of diverse species. Chromatin modification profiles are frequently noisy and diffuse, spanning regions ranging from several nucleosomes to large domains of multiple genes. Much of the early work on the identification of ChIP-enriched regions for ChIP-Seq data has focused on identifying localized regions, such as transcription factor binding sites. Bioinformatic tools to identify diffuse domains of ChIP-enriched regions have been lacking. Based on the biological observation that histone modifications tend to cluster to form domains, we present a method that identifies spatial clusters of signals unlikely to appear by chance. This method pools together enrichment information from neighboring nucleosomes to increase sensitivity and specificity. By using genomic-scale analysis, as well as the examination of loci with validated epigenetic states, we demonstrate that this method outperforms existing methods in the identification of ChIP-enriched signals for histone modification profiles. We demonstrate the application of this unbiased method in important issues in ChIP-Seq data analysis, such as data normalization for quantitative comparison of levels of epigenetic modifications across cell types and growth conditions. http://home.gwu.edu/ approximately wpeng/Software.htm. Supplementary data are available at Bioinformatics online.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Chromosome-wide and promoter-specific analyses identify sites of differential DNA methylation in normal and transformed human cells.

            Cytosine methylation is required for mammalian development and is often perturbed in human cancer. To determine how this epigenetic modification is distributed in the genomes of primary and transformed cells, we used an immunocapturing approach followed by DNA microarray analysis to generate methylation profiles of all human chromosomes at 80-kb resolution and for a large set of CpG islands. In primary cells we identified broad genomic regions of differential methylation with higher levels in gene-rich neighborhoods. Female and male cells had indistinguishable profiles for autosomes but differences on the X chromosome. The inactive X chromosome (Xi) was hypermethylated at only a subset of gene-rich regions and, unexpectedly, overall hypomethylated relative to its active counterpart. The chromosomal methylation profile of transformed cells was similar to that of primary cells. Nevertheless, we detected large genomic segments with hypomethylation in the transformed cell residing in gene-poor areas. Furthermore, analysis of 6,000 CpG islands showed that only a small set of promoters was methylated differentially, suggesting that aberrant methylation of CpG island promoters in malignancy might be less frequent than previously hypothesized.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes.

              Histone acetyltransferases (HATs) and deacetylases (HDACs) function antagonistically to control histone acetylation. As acetylation is a histone mark for active transcription, HATs have been associated with active and HDACs with inactive genes. We describe here genome-wide mapping of HATs and HDACs binding on chromatin and find that both are found at active genes with acetylated histones. Our data provide evidence that HATs and HDACs are both targeted to transcribed regions of active genes by phosphorylated RNA Pol II. Furthermore, the majority of HDACs in the human genome function to reset chromatin by removing acetylation at active genes. Inactive genes that are primed by MLL-mediated histone H3K4 methylation are subject to a dynamic cycle of acetylation and deacetylation by transient HAT/HDAC binding, preventing Pol II from binding to these genes but poising them for future activation. Silent genes without any H3K4 methylation signal show no evidence of being bound by HDACs.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Genet
                PLoS Genet
                plos
                plosgen
                PLoS Genetics
                Public Library of Science (San Francisco, USA )
                1553-7390
                1553-7404
                December 2013
                December 2013
                26 December 2013
                : 9
                : 12
                : e1004028
                Affiliations
                [1 ]Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
                [2 ]Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
                [3 ]SRA International, Inc., Durham, North Carolina, United States of America
                [4 ]Department of Biochemistry and Molecular Biology, Georgia Health Sciences University, Augusta, Georgia, United States of America
                Medical Research Council, Human Genetics Unit, United Kingdom
                Author notes

                The authors have declared that no competing interests exist.

                Conceived and designed the experiments: TS AD HS PAW. Performed the experiments: TS AD HS. Analyzed the data: YD SAG DM RRS. Wrote the paper: TS YD SAG AD DM RRS HS PAW.

                [¤]

                Current address: Department of Biochemistry and Molecular Biology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America.

                Article
                PGENETICS-D-13-01724
                10.1371/journal.pgen.1004028
                3873231
                24385926
                69770220-eb52-472a-a088-cabda8e01420
                Copyright @ 2013

                This is an open-access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.

                History
                : 26 June 2013
                : 31 October 2013
                Page count
                Pages: 15
                Funding
                This work was supported in part by the Intramural Research Program of the National Institute of Environmental Health Sciences, NIH (Project number ZO1ES101965 to PAW) and by a grant from NIH (CA134304 to HS). DM and RRS were supported in whole or in part with federal funds from the National Institute of Environmental Health Sciences, under Delivery Order Number HHSN291200555547C, GSA Contract Number GS-00F-003L. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article

                Genetics
                Genetics

                Comments

                Comment on this article