3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The Chromatin Environment Around Interneuron Genes in Oligodendrocyte Precursor Cells and Their Potential for Interneuron Reprograming

      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

          Oligodendrocyte precursor cells (OPCs), also known as NG2 glia, arise from neural progenitor cells in the embryonic ganglionic eminences that also generate inhibitory neurons. They are ubiquitously distributed in the central nervous system, remain proliferative through life, and generate oligodendrocytes in both gray and white matter. OPCs exhibit some lineage plasticity, and attempts have been made to reprogram them into neurons, with varying degrees of success. However, little is known about how epigenetic mechanisms affect the ability of OPCs to undergo fate switch and whether OPCs have a unique chromatin environment around neuronal genes that might contribute to their lineage plasticity. Our bioinformatic analysis of histone posttranslational modifications at interneuron genes in OPCs revealed that OPCs had significantly fewer bivalent and repressive histone marks at interneuron genes compared to astrocytes or fibroblasts. Conversely, OPCs had a greater degree of deposition of active histone modifications at bivalently marked interneuron genes than other cell types, and this was correlated with higher expression levels of these genes in OPCs. Furthermore, a significantly higher proportion of interneuron genes in OPCs than in other cell types lacked the histone posttranslational modifications examined. These genes had a moderately high level of expression, suggesting that the “no mark” interneuron genes could be in a transcriptionally “poised” or “transitional” state. Thus, our findings suggest that OPCs have a unique histone code at their interneuron genes that may obviate the need for erasure of repressive marks during their fate switch to inhibitory neurons.

          Related collections

          Most cited references82

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

          Direct conversion of fibroblasts to functional neurons by defined factors

          Cellular differentiation and lineage commitment are considered robust and irreversible processes during development. Recent work has shown that mouse and human fibroblasts can be reprogrammed to a pluripotent state with a combination of four transcription factors. This raised the question of whether transcription factors could directly induce other defined somatic cell fates, and not only an undifferentiated state. We hypothesized that combinatorial expression of neural lineage-specific transcription factors could directly convert fibroblasts into neurons. Starting from a pool of nineteen candidate genes, we identified a combination of only three factors, Ascl1, Brn2, and Myt1l, that suffice to rapidly and efficiently convert mouse embryonic and postnatal fibroblasts into functional neurons in vitro. These induced neuronal (iN) cells express multiple neuron-specific proteins, generate action potentials, and form functional synapses. Generation of iN cells from non-neural lineages could have important implications for studies of neural development, neurological disease modeling, and regenerative medicine.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            A unique chromatin signature uncovers early developmental enhancers in humans.

            Cell-fate transitions involve the integration of genomic information encoded by regulatory elements, such as enhancers, with the cellular environment. However, identification of genomic sequences that control human embryonic development represents a formidable challenge. Here we show that in human embryonic stem cells (hESCs), unique chromatin signatures identify two distinct classes of genomic elements, both of which are marked by the presence of chromatin regulators p300 and BRG1, monomethylation of histone H3 at lysine 4 (H3K4me1), and low nucleosomal density. In addition, elements of the first class are distinguished by the acetylation of histone H3 at lysine 27 (H3K27ac), overlap with previously characterized hESC enhancers, and are located proximally to genes expressed in hESCs and the epiblast. In contrast, elements of the second class, which we term 'poised enhancers', are distinguished by the absence of H3K27ac, enrichment of histone H3 lysine 27 trimethylation (H3K27me3), and are linked to genes inactive in hESCs and instead are involved in orchestrating early steps in embryogenesis, such as gastrulation, mesoderm formation and neurulation. Consistent with the poised identity, during differentiation of hESCs to neuroepithelium, a neuroectoderm-specific subset of poised enhancers acquires a chromatin signature associated with active enhancers. When assayed in zebrafish embryos, poised enhancers are able to direct cell-type and stage-specific expression characteristic of their proximal developmental gene, even in the absence of sequence conservation in the fish genome. Our data demonstrate that early developmental enhancers are epigenetically pre-marked in hESCs and indicate an unappreciated role of H3K27me3 at distal regulatory elements. Moreover, the wealth of new regulatory sequences identified here provides an invaluable resource for studies and isolation of transient, rare cell populations representing early stages of human embryogenesis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain.

              Heterochromatin protein 1 (HP1) is localized at heterochromatin sites where it mediates gene silencing. The chromo domain of HP1 is necessary for both targeting and transcriptional repression. In the fission yeast Schizosaccharomyces pombe, the correct localization of Swi6 (the HP1 equivalent) depends on Clr4, a homologue of the mammalian SUV39H1 histone methylase. Both Clr4 and SUV39H1 methylate specifically lysine 9 of histone H3 (ref. 6). Here we show that HP1 can bind with high affinity to histone H3 methylated at lysine 9 but not at lysine 4. The chromo domain of HP1 is identified as its methyl-lysine-binding domain. A point mutation in the chromo domain, which destroys the gene silencing activity of HP1 in Drosophila, abolishes methyl-lysine-binding activity. Genetic and biochemical analysis in S. pombe shows that the methylase activity of Clr4 is necessary for the correct localization of Swi6 at centromeric heterochromatin and for gene silencing. These results provide a stepwise model for the formation of a transcriptionally silent heterochromatin: SUV39H1 places a 'methyl marker' on histone H3, which is then recognized by HP1 through its chromo domain. This model may also explain the stable inheritance of the heterochromatic state.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Neurosci
                Front Neurosci
                Front. Neurosci.
                Frontiers in Neuroscience
                Frontiers Media S.A.
                1662-4548
                1662-453X
                08 August 2019
                2019
                : 13
                : 829
                Affiliations
                [1] 1Department of Physiology and Neurobiology, University of Connecticut , Storrs, CT, United States
                [2] 2Connecticut Institute for Brain and Cognitive Sciences, University of Connecticut , Storrs, CT, United States
                [3] 3Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University , Cleveland, OH, United States
                [4] 4Computational Biology Core, University of Connecticut , Storrs, CT, United States
                [5] 5Advanced Science Research Center at the Graduate Center, Neuroscience Initiative, The City University of New York , New York, NY, United States
                [6] 6Department of Pediatrics, University of Cincinnati , Cincinnati, OH, United States
                [7] 7Department of Computer Science and Engineering, University of Connecticut , Storrs, CT, United States
                [8] 8Institute for Systems Genomics, University of Connecticut , Storrs, CT, United States
                Author notes

                Edited by: Annalisa Buffo, University of Turin, Italy

                Reviewed by: Fernando de Castro, Cajal Institute (CSIC), Spain; Ken Arai, Massachusetts General Hospital and Harvard Medical School, United States; Kaylene M. Young, University of Tasmania, Australia

                *Correspondence: Akiko Nishiyama, akiko.nishiyama@ 123456uconn.edu

                This article was submitted to Neurogenesis, a section of the journal Frontiers in Neuroscience

                Article
                10.3389/fnins.2019.00829
                6694778
                31440130
                1a6a0213-589f-448d-9715-e50643115460
                Copyright © 2019 Boshans, Factor, Singh, Liu, Zhao, Mandoiu, Lu, Casaccia, Tesar and Nishiyama.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 27 December 2018
                : 25 July 2019
                Page count
                Figures: 5, Tables: 1, Equations: 0, References: 120, Pages: 19, Words: 0
                Funding
                Funded by: National Institute of Neurological Disorders and Stroke 10.13039/100000065
                Award ID: R01 NS074870
                Award ID: R01 NS072427
                Award ID: R01 NS075243
                Award ID: R01 NS0042925
                Award ID: R37 NS0429245
                Funded by: Citizens United for Research in Epilepsy 10.13039/100002736
                Award ID: 414253
                Funded by: National Multiple Sclerosis Society 10.13039/100000890
                Award ID: RG1612-26501
                Categories
                Neuroscience
                Original Research

                Neurosciences
                ng2,oligodendrocyte,inhibitory neuron,reprograming,chromatin,histone post-translational modification

                Comments

                Comment on this article