The Role of Epigenetic Mechanisms in the Regulation of Gene Expression in the Nervous System

Neuroepigenetics is a newly emerging field in neurobiology that addresses the epigenetic mechanism of gene expression regulation in various postmitotic neurons, both over time and in response to environmental stimuli. In addition to its fundamental contribution to our understanding of basic neuronal physiology, alterations in these neuroepigenetic mechanisms have been recently linked to numerous neurodevelopmental, psychiatric, and neurodegenerative disorders. This article provides a selective review of the role of DNA and histone modifications in neuronal signal-induced gene expression regulation, plasticity, and survival and how targeting these mechanisms could advance the development of future therapies. In addition, we discuss a recent discovery on how double-strand breaks of genomic DNA mediate the rapid induction of activity-dependent gene expression in neurons.

[1]  Avi Ma'ayan,et al.  Polycomb repressive complex 2 (PRC2) silences genes responsible for neurodegeneration , 2016, Nature Neuroscience.

[2]  G. Ming,et al.  Epigenetic mechanisms in neurogenesis , 2016, Nature Reviews Neuroscience.

[3]  Oliver Hobert,et al.  A map of terminal regulators of neuronal identity in Caenorhabditis elegans , 2016, Wiley interdisciplinary reviews. Developmental biology.

[4]  James A. Swenberg,et al.  DNA methylation on N6-adenine in mammalian embryonic stem cells , 2016, Nature.

[5]  Benjamin P. C. Chen,et al.  Transcriptional elongation requires DNA break-induced signalling , 2015, Nature Communications.

[6]  M. Roobol Perspective: Enforce the clinical guidelines , 2015, Nature.

[7]  Z. Weng,et al.  The Role of H3K4me3 in Transcriptional Regulation Is Altered in Huntington’s Disease , 2015, PloS one.

[8]  D. Schübeler,et al.  Loss of Ezh2 promotes a midbrain-to-forebrain identity switch by direct gene derepression and Wnt-dependent regulation , 2015, BMC Biology.

[9]  H. Zoghbi,et al.  Reversal of phenotypes in MECP2 duplication mice using genetic rescue or antisense oligos , 2015, Nature.

[10]  Tae-Kyung Kim,et al.  Stimulus-specific combinatorial functionality of neuronal c-fos enhancers , 2015, Nature Neuroscience.

[11]  H. Bjornsson The Mendelian disorders of the epigenetic machinery , 2015, Genome research.

[12]  J. Sweatt,et al.  DNA methylation regulates neuronal glutamatergic synaptic scaling , 2015, Science Signaling.

[13]  Terrence J. Sejnowski,et al.  Epigenomic Signatures of Neuronal Diversity in the Mammalian Brain , 2015, Neuron.

[14]  Andreas R. Pfenning,et al.  Activity-Induced DNA Breaks Govern the Expression of Neuronal Early-Response Genes , 2015, Cell.

[15]  Giovanni Coppola,et al.  Tet3 regulates synaptic transmission and homeostatic plasticity via DNA oxidation and repair , 2015, Nature Neuroscience.

[16]  Wei Li,et al.  MeCP2 binds to non-CG methylated DNA as neurons mature, influencing transcription and the timing of onset for Rett syndrome , 2015, Proceedings of the National Academy of Sciences.

[17]  G. Ming,et al.  Role of Tet1 and 5-hydroxymethylcytosine in cocaine action , 2015, Nature Neuroscience.

[18]  R. Simon,et al.  Transcriptional Response of Polycomb Group Genes to Status Epilepticus in Mice is Modified by Prior Exposure to Epileptic Preconditioning , 2015, Front. Neurol..

[19]  A. Tarakhovsky,et al.  Coupling of T cell receptor specificity to natural killer T cell development by bivalent histone H3 methylation , 2015, The Journal of experimental medicine.

[20]  Harrison W. Gabel,et al.  Disruption of DNA methylation-dependent long gene repression in Rett syndrome , 2015, Nature.

[21]  A. Bird,et al.  Rett syndrome: a complex disorder with simple roots , 2015, Nature Reviews Genetics.

[22]  Jaehoon Shin,et al.  Decoding neural transcriptomes and epigenomes via high-throughput sequencing , 2014, Nature Neuroscience.

[23]  Tae-Kyung Kim,et al.  Enhancer RNA facilitates NELF release from immediate early genes. , 2014, Molecular cell.

[24]  L. Tsai,et al.  DNA Damage and Its Links to Neurodegeneration , 2014, Neuron.

[25]  O. Hobert,et al.  Maintenance of postmitotic neuronal cell identity , 2014, Nature Neuroscience.

[26]  J. Schuurs-Hoeijmakers,et al.  TDP2 protects transcription from abortive topoisomerase activity and is required for normal neural function , 2014, Nature Genetics.

[27]  Hao Wu,et al.  Reversing DNA Methylation: Mechanisms, Genomics, and Biological Functions , 2014, Cell.

[28]  Guoping Fan,et al.  Distribution, recognition and regulation of non-CpG methylation in the adult mammalian brain , 2013, Nature Neuroscience.

[29]  J. D. Macklis,et al.  Molecular logic of neocortical projection neuron specification, development and diversity , 2013, Nature Reviews Neuroscience.

[30]  David A. Orlando,et al.  Global transcriptional and translational repression in human-embryonic-stem-cell-derived Rett syndrome neurons. , 2013, Cell stem cell.

[31]  J. David Sweatt,et al.  TET1 Controls CNS 5-Methylcytosine Hydroxylation, Active DNA Demethylation, Gene Transcription, and Memory Formation , 2013, Neuron.

[32]  Li-Huei Tsai,et al.  Tet1 Is Critical for Neuronal Activity-Regulated Gene Expression and Memory Extinction , 2013, Neuron.

[33]  Matthew D. Schultz,et al.  Global Epigenomic Reconfiguration During Mammalian Brain Development , 2013, Science.

[34]  Zachary D. Smith,et al.  Tet1 regulates adult hippocampal neurogenesis and cognition. , 2013, Cell stem cell.

[35]  A. Bird,et al.  Rett syndrome mutations abolish the interaction of MeCP2 with the NCoR/SMRT co-repressor , 2013, Nature Neuroscience.

[36]  Anatol C. Kreitzer,et al.  Physiological Brain Activity Causes DNA Double Strand Breaks in Neurons — Exacerbation by Amyloid-β , 2013, Nature Neuroscience.

[37]  Antoine H. F. M. Peters,et al.  Ezh2 Orchestrates Topographic Migration and Connectivity of Mouse Precerebellar Neurons , 2013, Science.

[38]  N. Heintz,et al.  MeCP2 binds to 5hmc enriched within active genes and accessible chromatin in the nervous system , 2012, Cell.

[39]  A. Bird,et al.  Disease Modeling Using Embryonic Stem Cells: MeCP2 Regulates Nuclear Size and RNA Synthesis in Neurons , 2012, Stem cells.

[40]  Oliver Hobert,et al.  Regulation of terminal differentiation programs in the nervous system. , 2011, Annual review of cell and developmental biology.

[41]  G. Ming,et al.  Emerging roles of TET proteins and 5-hydroxymethylcytosines in active DNA demethylation and beyond , 2011, Cell cycle.

[42]  Madeleine P. Ball,et al.  Neuronal activity modifies DNA methylation landscape in the adult brain , 2011, Nature Neuroscience.

[43]  M. Greenberg,et al.  Neuronal activity-regulated gene transcription in synapse development and cognitive function. , 2011, Cold Spring Harbor perspectives in biology.

[44]  G. Ming,et al.  Hydroxylation of 5-Methylcytosine by TET1 Promotes Active DNA Demethylation in the Adult Brain , 2011, Cell.

[45]  Yi Zhang,et al.  Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification , 2010, Nature.

[46]  A. Tarakhovsky,et al.  Ezh2, the histone methyltransferase of PRC2, regulates the balance between self-renewal and differentiation in the cerebral cortex , 2010, Proceedings of the National Academy of Sciences.

[47]  G. Kreiman,et al.  Widespread transcription at neuronal activity-regulated enhancers , 2010, Nature.

[48]  Guoping Fan,et al.  Dnmt1 and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons , 2010, Nature Neuroscience.

[49]  Robert S. Illingworth,et al.  Neuronal MeCP2 is expressed at near histone-octamer levels and globally alters the chromatin state. , 2010, Molecular cell.

[50]  M. MacDonald,et al.  Huntingtin facilitates polycomb repressive complex 2 , 2009, Human molecular genetics.

[51]  M. Vidal,et al.  Polycomb Limits the Neurogenic Competence of Neural Precursor Cells to Promote Astrogenic Fate Transition , 2009, Neuron.

[52]  N. Heintz,et al.  The Nuclear DNA Base 5-Hydroxymethylcytosine Is Present in Purkinje Neurons and the Brain , 2009, Science.

[53]  G. Bates,et al.  A Large Number of Protein Expression Changes Occur Early in Life and Precede Phenotype Onset in a Mouse Model for Huntington Disease*S , 2009, Molecular & Cellular Proteomics.

[54]  Hei Sook Sul,et al.  A Role of DNA-PK for the Metabolic Gene Regulation in Response to Insulin , 2009, Cell.

[55]  G. Ming,et al.  Neuronal Activity–Induced Gadd45b Promotes Epigenetic DNA Demethylation and Adult Neurogenesis , 2009, Science.

[56]  G. Turrigiano The Self-Tuning Neuron: Synaptic Scaling of Excitatory Synapses , 2008, Cell.

[57]  T. Bestor,et al.  The Colorful History of Active DNA Demethylation , 2008, Cell.

[58]  Michael B. Stadler,et al.  Lineage-specific polycomb targets and de novo DNA methylation define restriction and potential of neuronal progenitors. , 2008, Molecular cell.

[59]  Stephen T. C. Wong,et al.  MeCP2, a Key Contributor to Neurological Disease, Activates and Represses Transcription , 2008, Science.

[60]  E. Kavalali,et al.  Activity-Dependent Suppression of Miniature Neurotransmission through the Regulation of DNA Methylation , 2008, The Journal of Neuroscience.

[61]  P. Arlotta,et al.  Neuronal subtype specification in the cerebral cortex , 2007, Nature Reviews Neuroscience.

[62]  Adrian Bird,et al.  Perceptions of epigenetics , 2007, Nature.

[63]  Dustin E. Schones,et al.  High-Resolution Profiling of Histone Methylations in the Human Genome , 2007, Cell.

[64]  A. Bird,et al.  Reversal of Neurological Defects in a Mouse Model of Rett Syndrome , 2007, Science.

[65]  M. Yaffe Faculty Opinions recommendation of A topoisomerase IIbeta-mediated dsDNA break required for regulated transcription. , 2006 .

[66]  I. Cobos,et al.  Cellular patterns of transcription factor expression in developing cortical interneurons. , 2006, Cerebral cortex.

[67]  C. Glass,et al.  A Topoisomerase IIß-Mediated dsDNA Break Required for Regulated Transcription , 2006, Science.

[68]  H. Zoghbi,et al.  Mild overexpression of MeCP2 causes a progressive neurological disorder in mice. , 2004, Human molecular genetics.

[69]  R. Jaenisch,et al.  Expression of MeCP2 in postmitotic neurons rescues Rett syndrome in mice. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[70]  Daisuke Hattori,et al.  DNA Methylation-Related Chromatin Remodeling in Activity-Dependent Bdnf Gene Regulation , 2003, Science.

[71]  A. Bird,et al.  Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals , 2003, Nature Genetics.

[72]  Brigitte Wild,et al.  Histone Methyltransferase Activity of a Drosophila Polycomb Group Repressor Complex , 2002, Cell.

[73]  Ira M. Hall,et al.  Regulation of Heterochromatic Silencing and Histone H3 Lysine-9 Methylation by RNAi , 2002, Science.

[74]  Wendy A. Bickmore,et al.  Spatial organization of active and inactive genes and noncoding DNA within chromosome territories , 2002, The Journal of cell biology.

[75]  D. Baulcombe,et al.  Spreading of RNA Targeting and DNA Methylation in RNA Silencing Requires Transcription of the Target Gene and a Putative RNA-Dependent RNA Polymerase Article, publication date, and citation information can be found at www.plantcell.org/cgi/doi/10.1105/tpc.010480. , 2002, The Plant Cell Online.

[76]  E. Kandel The Molecular Biology of Memory Storage: A Dialogue Between Genes and Synapses , 2001, Science.

[77]  C. Allis,et al.  Translating the Histone Code , 2001, Science.

[78]  R. Kingston,et al.  Mechanisms of transcriptional memory , 2001, Nature Reviews Molecular Cell Biology.

[79]  H. Zoghbi,et al.  Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2 , 1999, Nature Genetics.

[80]  Colin A. Johnson,et al.  Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex , 1998, Nature.

[81]  Matthias Merkenschlager,et al.  Association of Transcriptionally Silent Genes with Ikaros Complexes at Centromeric Heterochromatin , 1997, Cell.

[82]  A. Bird,et al.  MeCP2 Is a Transcriptional Repressor with Abundant Binding Sites in Genomic Chromatin , 1997, Cell.

[83]  M. Bear,et al.  Metaplasticity: the plasticity of synaptic plasticity , 1996, Trends in Neurosciences.

[84]  A. Bird,et al.  Dissection of the methyl-CpG binding domain from the chromosomal protein MeCP2. , 1993, Nucleic acids research.

[85]  Wang,et al.  DNA methylation on N 6 -adenine in mammalian embryonic stem cells , 2016 .

[86]  K. Kroll,et al.  The roles and regulation of Polycomb complexes in neural development , 2014, Cell and Tissue Research.

[87]  David R. Liu,et al.  Conversion of 5-Methylcytosine to 5- Hydroxymethylcytosine in Mammalian DNA by the MLL Partner TET1 , 2009 .

[88]  J. Sweatt,et al.  Covalent Modification of DNA Regulates Memory Formation , 2008, Neuron.

[89]  A. Bird,et al.  Oxidative damage to methyl-CpG sequences inhibits the binding of the methyl-CpG binding domain (MBD) of methyl-CpG binding protein 2 (MeCP2). , 2004, Nucleic acids research.

[90]  A. Bird DNA methylation patterns and epigenetic memory. , 2002, Genes & development.

[91]  P. Jeffrey,et al.  Regulation of Heterochromatic Silencing and Histone H 3 Lysine-9 Methylation by RNAi , 2002 .