17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      A Functional Switch of NuRD Chromatin Remodeling Complex Subunits Regulates Mouse Cortical Development

      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

          Histone modifications and chromatin remodeling represent universal mechanisms by which cells adapt their transcriptional response to rapidly changing environmental conditions. Extensive chromatin remodeling takes place during neuronal development, allowing the transition of pluripotent cells into differentiated neurons. Here, we report that the NuRD complex, which couples ATP-dependent chromatin remodeling with histone deacetylase activity, regulates mouse brain development. Subunit exchange of CHDs, the core ATPase subunits of the NuRD complex, is required for distinct aspects of cortical development. Whereas CHD4 promotes the early proliferation of progenitors, CHD5 facilitates neuronal migration and CHD3 ensures proper layer specification. Inhibition of each CHD leads to defects of neuronal differentiation and migration, which cannot be rescued by expressing heterologous CHDs. Finally, we demonstrate that NuRD complexes containing specific CHDs are recruited to regulatory elements and modulate the expression of genes essential for brain development.

          Graphical Abstract

          Highlights

          • The ATPases CHD3, CHD4, and CHD5 are mutually exclusive subunits of the NuRD complex

          • CHD3, CHD4, and CHD5 regulate distinct and non-redundant aspects of cortical development

          • Loss of each CHD leads to specific defects of neuronal proliferation and migration

          • CHD3, CHD4, and CHD5 regulate distinct set of genes essential for brain development

          Abstract

          Neural development requires active chromatin remodeling. Nitarska et al. identify distinct NuRD chromatin remodeling complexes, containing CHD3, CHD4, or CHD5, during mouse embryonic cortical development. CHD4 promotes proliferation of basal progenitors, while CHD5 facilitates early radial migration and CHD3 drives late migration and laminar specification of neurons.

          Related collections

          Most cited references48

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

          Neuronal subtype-specific genes that control corticospinal motor neuron development in vivo.

          Within the vertebrate nervous system, the presence of many different lineages of neurons and glia complicates the molecular characterization of single neuronal populations. In order to elucidate molecular mechanisms underlying the specification and development of corticospinal motor neurons (CSMN), we purified CSMN at distinct stages of development in vivo and compared their gene expression to two other pure populations of cortical projection neurons: callosal projection neurons and corticotectal projection neurons. We found genes that are potentially instructive for CSMN development, as well as genes that are excluded from CSMN and are restricted to other populations of neurons, even within the same cortical layer. Loss-of-function experiments in null mutant mice for Ctip2 (also known as Bcl11b), one of the newly characterized genes, demonstrate that it plays a critical role in the development of CSMN axonal projections to the spinal cord in vivo, confirming that we identified central genetic determinants of the CSMN population.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Neural progenitors, neurogenesis and the evolution of the neocortex.

            The neocortex is the seat of higher cognitive functions and, in evolutionary terms, is the youngest part of the mammalian brain. Since its origin, the neocortex has expanded in several mammalian lineages, and this is particularly notable in humans. This expansion reflects an increase in the number of neocortical neurons, which is determined during development and primarily reflects the number of neurogenic divisions of distinct classes of neural progenitor cells. Consequently, the evolutionary expansion of the neocortex and the concomitant increase in the numbers of neurons produced during development entail interspecies differences in neural progenitor biology. Here, we review the diversity of neocortical neural progenitors, their interspecies variations and their roles in determining the evolutionary increase in neuron numbers and neocortex size. © 2014. Published by The Company of Biologists Ltd.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Satb2 is a postmitotic determinant for upper-layer neuron specification in the neocortex.

              Pyramidal neurons of the neocortex can be subdivided into two major groups: deep- (DL) and upper-layer (UL) neurons. Here we report that the expression of the AT-rich DNA-binding protein Satb2 defines two subclasses of UL neurons: UL1 (Satb2 positive) and UL2 (Satb2 negative). In the absence of Satb2, UL1 neurons lose their identity and activate DL- and UL2-specific genetic programs. UL1 neurons in Satb2 mutants fail to migrate to superficial layers and do not contribute to the corpus callosum but to the corticospinal tract, which is normally populated by DL axons. Ctip2, a gene required for the formation of the corticospinal tract, is ectopically expressed in all UL1 neurons in the absence of Satb2. Satb2 protein interacts with the Ctip2 genomic region and controls chromatin remodeling at this locus. Satb2 therefore is required for the initiation of the UL1-specific genetic program and for the inactivation of DL- and UL2-specific genes.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Rep
                Cell Rep
                Cell Reports
                Cell Press
                2211-1247
                01 November 2016
                01 November 2016
                01 November 2016
                : 17
                : 6
                : 1683-1698
                Affiliations
                [1 ]MRC Laboratory for Molecular and Cell Biology, University College London, London WC1E 6BT, UK
                [2 ]Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1737 USA
                [3 ]Lincoln’s Inn Fields Laboratory, The Francis Crick Institute, London WC2A 3LY, UK
                Author notes
                []Corresponding author a.riccio@ 123456ucl.ac.uk
                [4]

                Present address: The Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA

                [5]

                Lead Contact

                Article
                S2211-1247(16)31408-5
                10.1016/j.celrep.2016.10.022
                5149529
                27806305
                83d280c1-18ff-430e-ace1-c95522b00b48
                © 2016 The Author(s)

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

                History
                : 7 January 2016
                : 3 August 2016
                : 7 October 2016
                Categories
                Article

                Cell biology
                epigenetics,chromatin remodeling,chd proteins,nurd complex,mouse brain,cortical development,neural radial migration,neural progenitors,cortical laminar fate specification

                Comments

                Comment on this article