436
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      MERVL/Zscan4 Network Activation Results in Transient Genome-wide DNA Demethylation of mESCs

      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.

          Summary

          Mouse embryonic stem cells are dynamic and heterogeneous. For example, rare cells cycle through a state characterized by decondensed chromatin and expression of transcripts, including the Zscan4 cluster and MERVL endogenous retrovirus, which are usually restricted to preimplantation embryos. Here, we further characterize the dynamics and consequences of this transient cell state. Single-cell transcriptomics identified the earliest upregulated transcripts as cells enter the MERVL/Zscan4 state. The MERVL/Zscan4 transcriptional network was also upregulated during induced pluripotent stem cell reprogramming. Genome-wide DNA methylation and chromatin analyses revealed global DNA hypomethylation accompanying increased chromatin accessibility. This transient DNA demethylation was driven by a loss of DNA methyltransferase proteins in the cells and occurred genome-wide. While methylation levels were restored once cells exit this state, genomic imprints remained hypomethylated, demonstrating a potential global and enduring influence of endogenous retroviral activation on the epigenome.

          Graphical Abstract

          Highlights

          • Single-cell transcriptomics reveals dynamics of MERVL/Zscan4 network activation

          • MERVL-LTR transcriptional network is expressed in iPSC reprogramming events

          • Translation block depletes Dnmt proteins, inducing transient global demethylation

          • Passage through the MERVL/Zscan4 state may cause irreversible imprint erasure

          Abstract

          Mouse embryonic stem cells sporadically express preimplantation transcripts, including the MERVL endogenous retrovirus and Zscan4 cluster. Eckersley-Maslin et al. investigate the transcriptional dynamics in these cells and reveal transient genome-wide DNA demethylation accompanying chromatin decompaction. Following state exit, methylation levels are restored, except for genomic imprints, which remain lost.

          Related collections

          Most cited references30

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

          Genomic imprinting in mammals.

          Genomic imprinting affects a subset of genes in mammals and results in a monoallelic, parental-specific expression pattern. Most of these genes are located in clusters that are regulated through the use of insulators or long noncoding RNAs (lncRNAs). To distinguish the parental alleles, imprinted genes are epigenetically marked in gametes at imprinting control elements through the use of DNA methylation at the very least. Imprinted gene expression is subsequently conferred through lncRNAs, histone modifications, insulators, and higher-order chromatin structure. Such imprints are maintained after fertilization through these mechanisms despite extensive reprogramming of the mammalian genome. Genomic imprinting is an excellent model for understanding mammalian epigenetic regulation.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Single Cell RNA-Sequencing of Pluripotent States Unlocks Modular Transcriptional Variation

            Summary Embryonic stem cell (ESC) culture conditions are important for maintaining long-term self-renewal, and they influence cellular pluripotency state. Here, we report single cell RNA-sequencing of mESCs cultured in three different conditions: serum, 2i, and the alternative ground state a2i. We find that the cellular transcriptomes of cells grown in these conditions are distinct, with 2i being the most similar to blastocyst cells and including a subpopulation resembling the two-cell embryo state. Overall levels of intercellular gene expression heterogeneity are comparable across the three conditions. However, this masks variable expression of pluripotency genes in serum cells and homogeneous expression in 2i and a2i cells. Additionally, genes related to the cell cycle are more variably expressed in the 2i and a2i conditions. Mining of our dataset for correlations in gene expression allowed us to identify additional components of the pluripotency network, including Ptma and Zfp640, illustrating its value as a resource for future discovery.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Competition between DNA methylation and transcription factors determines binding of NRF1.

              Eukaryotic transcription factors (TFs) are key determinants of gene activity, yet they bind only a fraction of their corresponding DNA sequence motifs in any given cell type. Chromatin has the potential to restrict accessibility of binding sites; however, in which context chromatin states are instructive for TF binding remains mainly unknown. To explore the contribution of DNA methylation to constrained TF binding, we mapped DNase-I-hypersensitive sites in murine stem cells in the presence and absence of DNA methylation. Methylation-restricted sites are enriched for TF motifs containing CpGs, especially for those of NRF1. In fact, the TF NRF1 occupies several thousand additional sites in the unmethylated genome, resulting in increased transcription. Restoring de novo methyltransferase activity initiates remethylation at these sites and outcompetes NRF1 binding. This suggests that binding of DNA-methylation-sensitive TFs relies on additional determinants to induce local hypomethylation. In support of this model, removal of neighbouring motifs in cis or of a TF in trans causes local hypermethylation and subsequent loss of NRF1 binding. This competition between DNA methylation and TFs in vivo reveals a case of cooperativity between TFs that acts indirectly via DNA methylation. Methylation removal by methylation-insensitive factors enables occupancy of methylation-sensitive factors, a principle that rationalizes hypomethylation of regulatory regions.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Rep
                Cell Rep
                Cell Reports
                Cell Press
                2211-1247
                27 September 2016
                27 September 2016
                27 September 2016
                : 17
                : 1
                : 179-192
                Affiliations
                [1 ]Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK
                [2 ]EMBL-European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
                [3 ]Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
                [4 ]Laboratory of EpiGenetics, Saarland University, Campus A2 4, 66123 Saarbrücken, Germany
                [5 ]Bioinformatics Group, Babraham Institute, Cambridge CB22 3AQ, UK
                [6 ]Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
                [7 ]Computer Science Department, Saarland University, Campus E1.3, 66123 Saarbrücken, Germany
                Author notes
                []Corresponding author eckersleym@ 123456babraham.ac.uk
                [∗∗ ]Corresponding author wolf.reik@ 123456babraham.ac.uk
                [8]

                Lead Contact

                Article
                S2211-1247(16)31196-2
                10.1016/j.celrep.2016.08.087
                5055476
                27681430
                3ca0405b-09d0-4ba6-b032-22b1a85063e4
                © 2016 The Author(s)

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 3 June 2016
                : 19 July 2016
                : 25 August 2016
                Categories
                Article

                Cell biology
                zscan4,mervl,endogenous retrovirus,embryonic stem cell,dna methylation,imprint,preimplantation,reprogramming,chromatin

                Comments

                Comment on this article