Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Epigenetic regulation in psychiatric disorders

An Author Correction to this article was published on 13 December 2018

This article has been updated

Key Points

  • Epigenetic regulation of a gene is the process by which the activity of a particular gene is controlled by the structure of nearby chromatin.

  • Chromatin remodelling is complex and involves covalent modification of histones (for example, acetylation, methylation and phosphorylation), ATPase-containing protein complexes that move histone oligomers along a strand of DNA, direct methylation of DNA, and the binding of numerous transcription factors and transcriptional co-activators and co-repressors, all of which act in a concerted fashion to determine the activity of a given gene.

  • Epigenetic regulation is crucial for nervous system development, and several common mental retardation syndromes and related neurodevelopmental disorders are caused by abnormalities in chromatin remodelling mechanisms.

  • Epigenetic regulation also occurs in the mature brain, and may underlie stable changes in gene expression both under normal conditions (for example, learning and memory) and in several neuropathological states.

  • Long-lasting changes in histone acetylation, histone methylation and DNA methylation have been demonstrated in rodent models of depression. Drugs that increase histone acetylation exert antidepressant-like effects in these models.

  • Some of the lasting effects on the brain of drugs of abuse such as cocaine have been related to the drug's regulation of histone acetylation. Agents that increase histone acetylation enhance biochemical and behavioural responses to cocaine, and mice lacking certain enzymes that mediate histone deacetylation show similar increases in cocaine responsiveness.

  • Rett syndrome, an autism spectrum disorder, is caused by loss of function mutations in the gene that encodes a protein that binds to methylated sites in DNA and acts to repress the associated genes.

  • Recent work has implicated abnormalities in DNA methylation and histone acetylation in schizophrenia.

  • Work on epigenetic mechanisms of psychiatric disorders is in its early stages, but promises to improve our understanding of disease pathophysiology and might lead to the development of fundamentally new treatments for these conditions.

Abstract

Many neurological and most psychiatric disorders are not due to mutations in a single gene; rather, they involve molecular disturbances entailing multiple genes and signals that control their expression. Recent research has demonstrated that complex 'epigenetic' mechanisms, which regulate gene activity without altering the DNA code, have long-lasting effects within mature neurons. This review summarizes recent evidence for the existence of sustained epigenetic mechanisms of gene regulation in neurons that have been implicated in the regulation of complex behaviour, including abnormalities in several psychiatric disorders such as depression, drug addiction and schizophrenia.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: General scheme of chromatin remodelling.
Figure 2: Regulation of chromatin remodelling via intracellular signalling cascades.
Figure 3: Regulation of the Bdnf gene by social defeat.
Figure 4: Proposed chromatin remodelling events at a cocaine-activated gene.

Similar content being viewed by others

Change history

  • 14 December 2018

    In part c of Figure 1 in this article, the orientation of the tail of histone 3 with respect to the core region of this molecule was incorrect, and the amino acid residue K79 should not have been depicted in the tail. The corrected figure is shown below. The authors and editors thank T. Brown, R. Lober and C. Waker for bringing this error to our attention. Note that the present affiliation of the corresponding author and their current email address have been added to the correction notice.

References

  1. Kendler, K. S. Twin studies of psychiatric illness: an update. Arch. Gen. Psychiatry 58, 1005–1014 (2001).

    CAS  PubMed  Google Scholar 

  2. Hyman, S. E. & Nestler, E. J. The Molecular Foundations of Psychiatry (American Psychiatric, Washington, D. C., 1993).

    Google Scholar 

  3. McClung, C. A. et al. ΔFosB: a molecular switch for long-term adaptation in the brain. Brain Res. Mol. Brain Res. 132, 146–154 (2004).

    CAS  PubMed  Google Scholar 

  4. Nestler, E. J., Barrot, M. & Self, D. W. ΔFosB: a sustained molecular switch for addiction. Proc. Natl Acad. Sci. USA 98, 11042–11046 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Felsenfeld, G. & Groudine, M. Controlling the double helix. Nature 421, 448–453 (2003).

    PubMed  Google Scholar 

  6. Hake, S. B., Xiao, A. & Allis, C. D. Linking the epigenetic 'language' of covalent histone modifications to cancer. Br. J. Cancer 90, 761–769 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Lachner, M. & Jenuwein, T. The many faces of histone lysine methylation. Curr. Opin. Cell Biol. 14, 286–298 (2002).

    CAS  PubMed  Google Scholar 

  8. Gill, G. SUMO and ubiquitin in the nucleus: different functions, similar mechanisms? Genes Dev. 18, 2046–2059 (2004).

    CAS  PubMed  Google Scholar 

  9. Hassa, P. O., Haenni, S. S., Elser, M. & Hottiger, M. O. Nuclear ADP-ribosylation reactions in mammalian cells: where are we today and where are we going? Microbiol. Mol. Biol. Rev. 70, 789–829 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Jenuwein, T. & Allis, C. D. Translating the histone code. Science 293, 1074–1080 (2001).

    CAS  PubMed  Google Scholar 

  11. Narlikar, G. J., Fan, H. Y. & Kingston, R. E. Cooperation between complexes that regulate chromatin structure and transcription. Cell 108, 475–487 (2002).

    CAS  PubMed  Google Scholar 

  12. Shi, Y. et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941–953 (2004).

    CAS  PubMed  Google Scholar 

  13. Brami-Cherrier, K. et al. Parsing molecular and behavioral effects of cocaine in mitogen- and stress-activated protein kinase-1-deficient mice. J. Neurosci. 25, 11444–11454 (2005). Explores the signal transduction cascades, and their effect on downstream chromatin remodelling and associated gene expression, in striatal neurons in response to cocaine. It shows that cocaine causes induction of H4 acetylation, H3 phosphorylation and CREB phosphorylation through MSK1.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Kumar, A. et al. Chromatin remodeling is a key mechanism underlying cocaine-induced plasticity in striatum. Neuron 48, 303–314 (2005). Establishes an important role for chromatin remodelling in the reward responses to cocaine. Also, using chromatin immunoprecipitation assays, it shows that cocaine induces distinct histone modifications and in vivo binding of the transcription factor ΔFOSB at specific gene promoters in the striatum.

    CAS  PubMed  Google Scholar 

  15. Li, J. et al. Dopamine D2-like antagonists induce chromatin remodeling in striatal neurons through cyclic AMP-protein kinase A and NMDA receptor signaling. J. Neurochem. 90, 1117–1131 (2004). Demonstrates that acute administration of antipsychotic drugs to rodents increases global levels of histone acetylation in the striatum and provides evidence for the signal transduction mechanisms that mediate this effect.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Crosio, C., Heitz, E., Allis, C. D., Borrelli, E. & Sassone-Corsi, P. Chromatin remodeling and neuronal response: multiple signaling pathways induce specific histone H3 modifications and early gene expression in hippocampal neurons. J. Cell Sci. 116, 4905–4914 (2003).

    CAS  PubMed  Google Scholar 

  17. Bode, A. M. & Dong, Z. Inducible covalent posttranslational modification of histone H3. Sci. STKE 2005, re4 (2005).

    PubMed  Google Scholar 

  18. Chawla, S., Vanhoutte, P., Arnold, F. J., Huang, C. L. & Bading, H. Neuronal activity-dependent nucleocytoplasmic shuttling of HDAC4 and HDAC5. J. Neurochem. 85, 151–159 (2003).

    CAS  PubMed  Google Scholar 

  19. Linseman, D. A. et al. Inactivation of the myocyte enhancer factor-2 repressor histone deacetylase-5 by endogenous Ca2+ //calmodulin-dependent kinase II promotes depolarization-mediated cerebellar granule neuron survival. J. Biol. Chem. 278, 41472–41481 (2003).

    CAS  PubMed  Google Scholar 

  20. Gregoire, S. et al. Control of MEF2 transcriptional activity by coordinated phosphorylation and sumoylation. J. Biol. Chem. 281, 4423–4433 (2006).

    CAS  PubMed  Google Scholar 

  21. Berton, O. & Nestler, E. J. New approaches to antidepressant drug discovery: beyond monoamines. Nature Rev. Neurosci. 7, 137–151 (2006).

    CAS  Google Scholar 

  22. Tsankova, N. M., Kumar, A. & Nestler, E. J. Histone modifications at gene promoter regions in rat hippocampus after acute and chronic electroconvulsive seizures. J. Neurosci. 24, 5603–5610 (2004). Outlines a standardized approach to performing chromatin immunoprecipitation assays in rodent brain tissue. It also provides a detailed analysis of several transient and lasting changes in histone modifications after acute and chronic seizure, in correlation with changes in gene expression at the specific gene promoters.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Duman, R. S. Depression: a case of neuronal life and death? Biol. Psychiatry 56, 140–145 (2004).

    PubMed  Google Scholar 

  24. Nestler, E. J. et al. Neurobiology of depression. Neuron 34, 13–25 (2002).

    CAS  PubMed  Google Scholar 

  25. Duman, R. S. Role of neurotrophic factors in the etiology and treatment of mood disorders. Neuromolecular Med. 5, 11–25 (2004).

    CAS  PubMed  Google Scholar 

  26. Monteggia, L. M. et al. Essential role of brain-derived neurotrophic factor in adult hippocampal function. Proc. Natl Acad. Sci. USA 101, 10827–10832 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Monteggia, L. M. et al. Brain-derived neurotrophic factor conditional knockouts show gender differences in depression-related behaviors. Biol. Psychiatry 61, 187–197 (2006).

    PubMed  Google Scholar 

  28. Berton, O. et al. Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science 311, 864–868 (2006). The authors use chronic social defeat stress as an animal model of depression and demonstrate a crucial role for the neurotrophic factor BDNF in the mesolimbic dopamine pathway in mediating some of the deleterious molecular and behavioural sequelae of this stress paradigm.

    CAS  PubMed  Google Scholar 

  29. Tsankova, N. M. et al. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nature Neurosci. 9, 519–525 (2006). The first study to examine the involvement of chromatin remodelling in an animal model of depression. It reveals robust and lasting in vivo changes in histone modifications, and a role for HDAC5 in chronic social defeat stress and in antidepressant efficacy.

    CAS  PubMed  Google Scholar 

  30. Schroeder, F. A., Lin, C. L., Crusio, W. E. & Akbarian, S. Antidepressant-like effects of the histone deacetylase inhibitor, sodium butyrate, in the mouse (in the press).

  31. Lee, M. G., Wynder, C., Schmidt, D. M., McCafferty, D. G. & Shiekhattar, R. Histone H3 lysine 4 demethylation is a target of nonselective antidepressive medications. Chem. Biol. 13, 563–567 (2006).

    CAS  PubMed  Google Scholar 

  32. Champagne, F. A., Francis, D. D., Mar, A. & Meaney, M. J. Variations in maternal care in the rat as a mediating influence for the effects of environment on development. Physiol. Behav. 79, 359–371 (2003).

    CAS  PubMed  Google Scholar 

  33. Meaney, M. J. & Szyf, M. Maternal care as a model for experience-dependent chromatin plasticity? Trends Neurosci. 28, 456–463 (2005).

    CAS  PubMed  Google Scholar 

  34. Weaver, I. C. et al. Epigenetic programming by maternal behavior. Nature Neurosci. 7, 847–854 (2004). This important study provides highly novel evidence that the epigenetic state of GR in the hippocampus of rodent offspring, in particular its level of DNA methylation, can be modulated by maternal nurturing behavior in a lasting but reversible manner.

    CAS  PubMed  Google Scholar 

  35. Levenson, J. M. et al. Evidence that DNA (cytosine-5) methyltransferase regulates synaptic plasticity in the hippocampus. J. Biol. Chem. 281, 15763–15773 (2006). Along with reference 69, this study implicates rapid and reversible changes in DNA methylation in synaptic plasticity in the rodent hippocampus, and in the formation of long-term memory. The notion that DNA methylation is subject to dynamic regulation in the adult brain is highly novel and has important implications for our understanding of the epigenetic control of brain function.

    CAS  PubMed  Google Scholar 

  36. Everitt, B. J. & Robbins, T. W. Neural systems of reinforcement for drug addiction: from actions to habits to compulsion. Nature Neurosci. 8, 1481–1489 (2005).

    CAS  PubMed  Google Scholar 

  37. Hyman, S. E., Malenka, R. C. & Nestler, E. J. Neural mechanisms of addiction: the role of reward-related learning and memory. Annu. Rev. Neurosci. 29, 565–598 (2006).

    CAS  PubMed  Google Scholar 

  38. Freeman, W. M. et al. Cocaine-responsive gene expression changes in rat hippocampus. Neuroscience 108, 371–380 (2001).

    CAS  PubMed  Google Scholar 

  39. Freeman, W. M. et al. Changes in rat frontal cortex gene expression following chronic cocaine. Brain Res. Mol. Brain Res. 104, 11–20 (2002).

    CAS  PubMed  Google Scholar 

  40. Kreek, M. J., Bart, G., Lilly, C., LaForge, K. S. & Nielsen, D. A. Pharmacogenetics and human molecular genetics of opiate and cocaine addictions and their treatments. Pharmacol. Rev. 57, 1–26 (2005).

    CAS  PubMed  Google Scholar 

  41. McClung, C. A. & Nestler, E. J. Regulation of gene expression and cocaine reward by CREB and ΔFosB. Nature Neurosci. 6, 1208–1215 (2003).

    CAS  PubMed  Google Scholar 

  42. McClung, C. A. et al. Regulation of gene expression by chronic morphine and morphine withdrawal in the locus ceruleus and ventral tegmental area. J. Neurosci. 25, 6005–6015 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Yao, W. D. et al. Identification of PSD-95 as a regulator of dopamine-mediated synaptic and behavioral plasticity. Neuron 41, 625–638 (2004).

    CAS  PubMed  Google Scholar 

  44. Grimm, J. W. et al. Time-dependent increases in brain-derived neurotrophic factor protein levels within the mesolimbic dopamine system after withdrawal from cocaine: implications for incubation of cocaine craving. J. Neurosci. 23, 742–747 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Colvis, C. M. et al. Epigenetic mechanisms and gene networks in the nervous system. J. Neurosci. 25, 10379–10389 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Levine, A. A. et al. CREB-binding protein controls response to cocaine by acetylating histones at the fosB promoter in the mouse striatum. Proc. Natl Acad. Sci. USA 102, 19186–19191 (2005). Characterizes the influence of chromatin remodelling on cocaine action in the brain. In particular, it shows that recruitment of CBP to the FosB promoter and the resulting H4 acetylation are essential for normal levels of FosB expression, for accumulation of the transcription factor ΔFOSB, and for normal sensitivity to cocaine.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Bibb, J. A. et al. Effects of chronic exposure to cocaine are regulated by the neuronal protein Cdk5. Nature 410, 376–380 (2001).

    CAS  PubMed  Google Scholar 

  48. Lee, M. P. Genome-wide analysis of epigenetics in cancer. Ann. NY Acad. Sci. 983, 101–109 (2003).

    CAS  PubMed  Google Scholar 

  49. Lee, T. I. et al. Control of developmental regulators by Polycomb in human embryonic stem cells. Cell 125, 301–313 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Impey, S. et al. Defining the CREB regulon: a genome-wide analysis of transcription factor regulatory regions. Cell 119, 1041–1054 (2004). Introduces a novel method of genome-wide analysis of transcription factor binding sites, termed SACO, which combines chromatin immunoprecipitation with long serial analysis of gene expression by direct sequencing rather than by the use of microarray chips.

    CAS  PubMed  Google Scholar 

  51. Kumar, A. et al. Global maps of histone acetylation and gene regulatory networks in the nucleus accumbens after chronic cocaine using chip on chip. Soc. Neurosci. Abstr. 451.4 (2005).

  52. Kumar, A. et al. Transcriptional and post-transcriptional regulation of gene expression in nucleus accumbens associated with chronic stress-induced neuroadaptations in mouse. Soc. Neurosci. Abstr. 191.23 (2006).

  53. Renthal, W. et al. Epigenetic control of cocaine reward by class II histone deacetylases. Soc. Neurosci. Abstr. 294.27 (2006).

  54. Norrholm, S. D. et al. Cocaine-induced proliferation of dendritic spines in nucleus accumbens is dependent on the activity of cyclin-dependent kinase-5. Neuroscience 116, 19–22 (2003).

    CAS  PubMed  Google Scholar 

  55. Cassel, S. et al. Fluoxetine and cocaine induce the epigenetic factors MeCP2 and MBD1 in adult rat brain. Mol. Pharmacol. 70, 487–492 (2006).

    CAS  PubMed  Google Scholar 

  56. Li, Z. et al. Cdk5/p35 phosphorylates mSds3 and regulates mSds3-mediated repression of transcription. J. Biol. Chem. 279, 54438–54444 (2004).

    CAS  PubMed  Google Scholar 

  57. Korutla, L., Wang, P. J. & Mackler, S. A. The POZ/BTB protein NAC1 interacts with two different histone deacetylases in neuronal-like cultures. J. Neurochem. 94, 786–793 (2005).

    CAS  PubMed  Google Scholar 

  58. Mahadev, K. & Vemuri, M. C. Effect of ethanol on chromatin and nonhistone nuclear proteins in rat brain. Neurochem. Res. 23, 1179–1184 (1998).

    CAS  PubMed  Google Scholar 

  59. Bonsch, D., Lenz, B., Kornhuber, J. & Bleich, S. DNA hypermethylation of the alpha synuclein promoter in patients with alcoholism. Neuroreport 16, 167–170 (2005).

    PubMed  Google Scholar 

  60. Kim, J. S. & Shukla, S. D. Acute in vivo effect of ethanol (binge drinking) on histone H3 modifications in rat tissues. Alcohol Alcohol. 41, 126–132 (2006).

    CAS  PubMed  Google Scholar 

  61. Bailey, C. H., Kandel, E. R. & Si, K. The persistence of long-term memory: a molecular approach to self-sustaining changes in learning-induced synaptic growth. Neuron 44, 49–57 (2004).

    CAS  PubMed  Google Scholar 

  62. Levenson, J. M. & Sweatt, J. D. Epigenetic mechanisms in memory formation. Nature Rev. Neurosci. 6, 108–118 (2005).

    CAS  Google Scholar 

  63. Alarcon, J. M. et al. Chromatin acetylation, memory, and LTP are impaired in CBP+/− mice: a model for the cognitive deficit in Rubinstein–Taybi syndrome and its amelioration. Neuron 42, 947–959 (2004).

    CAS  PubMed  Google Scholar 

  64. Korzus, E., Rosenfeld, M. G. & Mayford, M. CBP histone acetyltransferase activity is a critical component of memory consolidation. Neuron 42, 961–972 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Levenson, J. M. et al. Regulation of histone acetylation during memory formation in the hippocampus. J. Biol. Chem. 279, 40545–40559 (2004).

    CAS  PubMed  Google Scholar 

  66. Chwang, W. B., O'Riordan, K. J., Levenson, J. M. & Sweatt, J. D. ERK/MAPK regulates hippocampal histone phosphorylation following contextual fear conditioning. Learn. Mem. 13, 322–328 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Kim, Y. et al. Epigenetic regulation of brain function by Polycomb and Trithorax complexes. Soc. Neurosci. Abstr. 750.5 (2006).

  68. Guan, Z. et al. Integration of long-term-memory-related synaptic plasticity involves bidirectional regulation of gene expression and chromatin structure. Cell 111, 483–493 (2002).

    CAS  PubMed  Google Scholar 

  69. Miller, C. A. & Sweatt, J. D. Covalent modification of DNA regulates memory formation. Neuron 53, 857–869 (2007). Demonstrates rapid changes in the methylation of several memory-related genes (for example, Bdnf, PP1 and reelin) in the hippocampus during contextual fear conditioning. The study, along with reference 35, provides one of the best indications so far that DNA methylation may be rapidly induced and reversed in the adult brain.

    CAS  PubMed  Google Scholar 

  70. Ausio, J., Levin, D. B., De Amorim, G. V., Bakker, S. & Macleod, P. M. Syndromes of disordered chromatin remodeling. Clin. Genet. 64, 83–95 (2003).

    CAS  PubMed  Google Scholar 

  71. Zoghbi, H. Y. MeCP2 dysfunction in humans and mice. J. Child Neurol. 20, 736–740 (2005).

    PubMed  Google Scholar 

  72. Amir, R. E. et al. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nature Genet. 23, 185–188 (1999).

    CAS  PubMed  Google Scholar 

  73. Luikenhuis, S., Giacometti, E., Beard, C. F. & Jaenisch, R. Expression of MeCP2 in postmitotic neurons rescues Rett syndrome in mice. Proc. Natl Acad. Sci. USA 101, 6033–6038 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Dani, V. S. et al. Reduced cortical activity due to a shift in the balance between excitation and inhibition in a mouse model of Rett syndrome. Proc. Natl Acad. Sci. USA 102, 12560–12565 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Chang, Q., Khare, G., Dani, V., Nelson, S. & Jaenisch, R. The disease progression of Mecp2 mutant mice is affected by the level of BDNF expression. Neuron 49, 341–348 (2006).

    CAS  PubMed  Google Scholar 

  76. Gemelli, T. et al. Postnatal loss of methyl-CpG binding protein 2 in the forebrain is sufficient to mediate behavioral aspects of Rett syndrome in mice. Biol. Psychiatry 59, 468–476 (2006).

    CAS  PubMed  Google Scholar 

  77. Moretti, P. et al. Learning and memory and synaptic plasticity are impaired in a mouse model of Rett syndrome. J. Neurosci. 26, 319–327 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Nelson, E. D., Kavalali, E. T. & Monteggia, L. M. MeCP2-dependent transcriptional repression regulates excitatory neurotransmission. Curr. Biol. 16, 710–716 (2006). Characterizes abnormalities in presynaptic excitatory transmission in cultured hippocampal neurons from mice lacking MeCP2. It shows that such abnormalities are not developmental in nature, but rather can be induced in adult neurons by deletion of Mecp2 and, conversely, that the consequences of early gene deletion can be corrected by inhibitors of transcription.

    CAS  PubMed  Google Scholar 

  79. Chen, R. Z., Akbarian, S., Tudor, M. & Jaenisch, R. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nature Genet. 27, 327–331 (2001).

    CAS  PubMed  Google Scholar 

  80. McGill, B. E. et al. Enhanced anxiety and stress-induced corticosterone release are associated with increased Crh expression in a mouse model of Rett syndrome. Proc. Natl Acad. Sci. USA 103, 18267–18272 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Guy, J., Gan, J., Selfridge, J., Cobb, S. & Bird, A. Reversal of neurological defects in a mouse model of Rett syndrome. Science 315, 1143–1147 (2007). Shows that Rett-like symptoms induced in mice as a result of loss of function mutations in MeCP2 can be largely reversed upon correction of the MeCP2 deficit. This supports the notion that it may be possible to treat Rett syndrome in humans even after it has become symptomatic.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Tremolizzo, L. et al. An epigenetic mouse model for molecular and behavioral neuropathologies related to schizophrenia vulnerability. Proc. Natl Acad. Sci. USA 99, 17095–17100 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Tamminga, C. A. & Holcomb, H. H. Phenotype of schizophrenia: a review and formulation. Mol. Psychiatry 10, 27–39 (2005).

    CAS  PubMed  Google Scholar 

  84. Grayson, D. R. et al. The human reelin gene: transcription factors (+), repressors (−) and the methylation switch (+/−) in schizophrenia. Pharmacol. Ther. 111, 272–286 (2006).

    CAS  PubMed  Google Scholar 

  85. Chen, Y., Sharma, R. P., Costa, R. H., Costa, E. & Grayson, D. R. On the epigenetic regulation of the human reelin promoter. Nucleic Acids Res. 30, 2930–2939 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Dong, E. et al. Reelin and glutamic acid decarboxylase67 promoter remodeling in an epigenetic methionine-induced mouse model of schizophrenia. Proc. Natl Acad. Sci. USA 102, 12578–12583 (2005). Provides evidence to support their hypothesis that epigenetic abnormalities, specifically, altered methylation of the reelin and Gad67 gene promoters, contribute to the pathophysiology of schizophrenia in a mouse model.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Antun, F. T. et al. The effects of L-methionine (without MAOI) in schizophrenia. J. Psychiatr. Res. 8, 63–71 (1971).

    CAS  PubMed  Google Scholar 

  88. Tremolizzo, L. et al. Valproate corrects the schizophrenia-like epigenetic behavioral modifications induced by methionine in mice. Biol. Psychiatry 57, 500–509 (2005).

    CAS  PubMed  Google Scholar 

  89. Cervoni, N. & Szyf, M. Demethylase activity is directed by histone acetylation. J. Biol. Chem. 276, 40778–84077 (2001).

    CAS  PubMed  Google Scholar 

  90. Cervoni, N., Detich, N., Seo, S. B., Chakravarti, D. & Szyf, M. The oncoprotein Set/TAF-1β, an inhibitor of histone acetyltransferase, inhibits active demethylation of DNA, integrating DNA methylation and transcriptional silencing. J. Biol. Chem. 277, 25026–25031 (2002).

    CAS  PubMed  Google Scholar 

  91. Casey, D. E. et al. Effect of divalproex combined with olanzapine or risperidone in patients with an acute exacerbation of schizophrenia. Neuropsychopharmacology 28, 182–192 (2003).

    CAS  PubMed  Google Scholar 

  92. Tsankov, A. M. et al. Communication between levels of transcriptional control improves robustness and adaptivity. Mol. Syst. Biol. 2, 65 (2006). Provides a system-level view of how transcription factors, chromatin regulators, RNA processing and nuclear transport proteins affect gene expression, revealing an elegant architecture for transcriptional control that improves the resilience and responsiveness of the eukaryotic cell.

    PubMed  PubMed Central  Google Scholar 

  93. Hsieh, J. & Gage, F. H. Chromatin remodeling in neural development and plasticity. Curr. Opin. Cell Biol. 17, 664–671 (2005).

    CAS  PubMed  Google Scholar 

  94. Ballas, N. & Mandel, G. The many faces of REST oversee epigenetic programming of neuronal genes. Curr. Opin. Neurobiol. 15, 500–506 (2005).

    CAS  PubMed  Google Scholar 

  95. Kuwabara, T., Hsieh, J., Nakashima, K., Taira, K. & Gage, F. H. A small modulatory dsRNA specifies the fate of adult neural stem cells. Cell 116, 779–793 (2004).

    CAS  PubMed  Google Scholar 

  96. Marin-Husstege, M., Muggironi, M., Liu, A. & Casaccia-Bonnefil, P. Histone deacetylase activity is necessary for oligodendrocyte lineage progression. J. Neurosci. 22, 10333–10345 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Fan, G. et al. DNA hypomethylation perturbs the function and survival of CNS neurons in postnatal animals. J. Neurosci. 21, 788–797 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Zhao, X. et al. Mice lacking methyl-CpG binding protein 1 have deficits in adult neurogenesis and hippocampal function. Proc. Natl Acad. Sci. USA 100, 6777–6782 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Liu, Q. R. et al. Rodent BDNF genes, novel promoters, novel splice variants, and regulation by cocaine. Brain Res. 1067, 1–12 (2006).

    CAS  PubMed  Google Scholar 

  100. Timmusk, T. et al. Multiple promoters direct tissue-specific expression of the rat BDNF gene. Neuron 10, 475–489 (1993).

    CAS  PubMed  Google Scholar 

  101. Tao, X., Finkbeiner, S., Arnold, D. B., Shaywitz, A. J. & Greenberg, M. E. Ca2+ influx regulates BDNF transcription by a CREB family transcription factor-dependent mechanism. Neuron 20, 709–726 (1998).

    CAS  PubMed  Google Scholar 

  102. Chen, W. G. et al. Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2. Science 302, 885–889 (2003).

    CAS  PubMed  Google Scholar 

  103. Zhou, Z. et al. Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation. Neuron 52, 255–269 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Huang, Y., Doherty, J. J. & Dingledine, R. Altered histone acetylation at glutamate receptor 2 and brain-derived neurotrophic factor genes is an early event triggered by status epilepticus. J. Neurosci. 22, 8422–8428 (2002). The first paper to demonstrate regulation of histone acetylation in the adult rat brain. The authors characterize rapid changes in acetylation at the Bdnf and Glur2 promoters in response to chemically induced seizures.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Lim, J. H., Booker, A. B. & Fallon, J. R. Regulating fragile X gene transcription in the brain and beyond. J. Cell Physiol. 205, 170–175 (2005).

    CAS  PubMed  Google Scholar 

  106. Merienne, K., Pannetier, S., Harel-Bellan, A. & Sassone-Corsi, P. Mitogen-regulated RSK2-CBP interaction controls their kinase and acetylase activities. Mol. Cell Biol. 21, 7089–7096 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Weeber, E. J., Levenson, J. M. & Sweatt, J. D. Molecular genetics of human cognition. Mol. Interv. 2, 376–91, 339 (2002).

    CAS  PubMed  Google Scholar 

  108. Davies, W., Isles, A. R. & Wilkinson, L. S. Imprinted gene expression in the brain. Neurosci. Biobehav. Rev. 29, 421–430 (2005).

    CAS  PubMed  Google Scholar 

  109. Vo, N. & Goodman, R. H. CREB-binding protein and p300 in transcriptional regulation. J. Biol. Chem. 276, 13505–13508 (2001).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Preparation of this review was supported by grants from the National Institute on Drug Abuse and the National Institute of Mental Health.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Eric J. Nestler.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information S1 (table)

Tsankova et al., Epigenetic Regulation in Psychiatric Disorders (PDF 215 kb)

Related links

Related links

DATABASES

OMIM

Rett syndrome

Rubinstein–Taybi syndrome

schizophrenia

FURTHER INFORMATION

Nestler's laboratory

Glossary

Histones

Highly basic proteins that comprise the major protein constituents of the nucleus. Octomeric complexes of histones, around which DNA is wrapped, form the nucleosome, the basic building block of chromatin.

Nucleosome

The basic building block of chromatin in which 147 base pairs of DNA are wrapped (1.65 turns) around a core histone octamer.

Heterochromatin

The inactivated state of chromatin, in which DNA is not accessible for transcription due to covalent modifications of histones, methylation of the DNA and the binding of numerous repressor proteins.

Euchromatin

The activated state of chromatin, in which sections of DNA are accessible to the transcriptional machinery.

Ubiquitylation

Covalent addition of a small protein, called ubiquitin, to many types of proteins. Addition of multiple ubiquitin groups, polyubiquitylation, targets proteins for degradation in the proteasome. By contrast, monoubiquitylation of histones and other regulatory proteins alters their functional properties.

SUMOylation

Covalent addition of SUMO, which are small ubiquitin-related modifier proteins, to histones and many other types of regulatory proteins, which alters those proteins' function.

Nucleosome sliding

The movement of the core histone octamer relative to the DNA, which allows that DNA to be progressively transcribed into RNA.

SWI/SNF

Protein complex that partly mediates nucleosome sliding in an ATP-dependent manner. The name comes from genetic screens of yeast which identified proteins implicated in mating switching and sucrose non-fermentation. The proteins were later found to regulate nucleosome sliding.

Histone substitution

A type of chromatin remodelling where histone constituents of the nucleosome can be replaced by other naturally occurring histone variants.

X-chromosome inactivation

Chromatin remodelling on a very large scale, whereby one of two X chromosomes in all cells of a female organism are inactivated by DNA hypermethylation. Once that chromosome is silenced, it remains inactive for the life of the organism.

Genetic imprinting

A process where only the maternal or paternal allele of a gene is expressed in the offspring. The other, inactivated allele is transcriptionally silenced through DNA methylation at CpG-rich domains.

Chromatin immunoprecipitation

(ChIP). A method that enables the identification of histone modifications or transcriptional regulatory proteins at a given gene promoter. In the assay, DNA is crosslinked to nearby proteins by light fixation, the material is sheared, then immunoprecipitated with an antibody to a particular protein of interest, and genes in the final immunoprecipitate are quantified by polymerase chain reaction.

Immediate-early genes

Genes that are induced rapidly and transiently without the need for new protein synthesis. Many immediate-early genes, such as c-Fos, control the transcription of other genes, and thereby provide the early stages in the control of the production of specific proteins.

DNA demethylases

Enzymes, not yet molecularly characterized, that demethylate CpG residues in DNA. Active DNA demethylation may also occur through alternative mechanisms such as DNA repair and deamination.

ChIP on chip

A method that enables a global analysis of genes associated with a particular histone modification or transcriptional regulatory protein. Immunoprecipitated chromatin is analysed on a microarray gene chip, enriched in promoter regions.

SACO

Serial analysis of chromatin occupancy, an alternative method to ChIP on chip, is used to obtain a genome-wide appreciation of the genes that bind a particular histone modification or transcriptional regulatory protein. Instead of hybridizing the immunoprecipitated DNA to a microarray, the DNA is directly sequenced.

Circular dichroism

A form of spectroscopy, involving the differential absorption of left- and right-handed polarized light, used to study the structure of complex molecules.

Valproate

A commonly used anticonvulsant and antimanic medication. Among many other actions (for example, direct effects on the brains GABA pathways), valproate is a nonspecific inhibitor of class I and class II HDACs.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Tsankova, N., Renthal, W., Kumar, A. et al. Epigenetic regulation in psychiatric disorders. Nat Rev Neurosci 8, 355–367 (2007). https://doi.org/10.1038/nrn2132

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn2132

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing