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.

  • Article
  • Published:

HDAC2 regulates atypical antipsychotic responses through the modulation of mGlu2 promoter activity

Abstract

Histone deacetylases (HDACs) compact chromatin structure and repress gene transcription. In schizophrenia, clinical studies demonstrate that HDAC inhibitors are efficacious when given in combination with atypical antipsychotics. However, the molecular mechanism that integrates a better response to antipsychotics with changes in chromatin structure remains unknown. Here we found that chronic atypical antipsychotics downregulated the transcription of metabotropic glutamate 2 receptor (mGlu2, also known as Grm2), an effect that was associated with decreased histone acetylation at its promoter in mouse and human frontal cortex. This epigenetic change occurred in concert with a serotonin 5-HT2A receptor–dependent upregulation and increased binding of HDAC2 to the mGlu2 promoter. Virally mediated overexpression of HDAC2 in frontal cortex decreased mGlu2 transcription and its electrophysiological properties, thereby increasing psychosis-like behavior. Conversely, HDAC inhibitors prevented the repressive histone modifications induced at the mGlu2 promoter by atypical antipsychotics, and augmented their therapeutic-like effects. These observations support the view of HDAC2 as a promising new target for schizophrenia treatment.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Decreased acetylation of histone H3 (H3ac) at the mGlu2 promoter by chronic treatment with atypical antipsychotic drugs in mouse frontal cortex.
Figure 2: Decreased acetylation of histone H3 (H3ac) at the mGlu2 promoter in prefrontal cortex of treated, but not untreated, subjects with schizophrenia.
Figure 3: 5-HT2A-dependent increased expression of HDAC2 in frontal cortex by chronic atypical antipsychotics.
Figure 4: HDAC2 binds to the mGlu2 promoter and represses its function.
Figure 5: HSV-HDAC2 in frontal cortex decreases mGlu2-dependent behavioral function.
Figure 6: Chronic treatment with SAHA prevents the repressive histone modifications induced at the mGlu2 promoter by chronic clozapine.

Similar content being viewed by others

References

  1. Sawa, A. & Snyder, S.H. Schizophrenia: diverse approaches to a complex disease. Science 296, 692–695 (2002).

    Article  CAS  Google Scholar 

  2. Dobbs, D. Schizophrenia: the making of a troubled mind. Nature 468, 154–156 (2010).

    Article  CAS  Google Scholar 

  3. Lieberman, J.A. et al. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N. Engl. J. Med. 353, 1209–1223 (2005).

    Article  CAS  Google Scholar 

  4. Lieberman, J.A. et al. Antipsychotic drugs: comparison in animal models of efficacy, neurotransmitter regulation, and neuroprotection. Pharmacol. Rev. 60, 358–403 (2008).

    Article  CAS  Google Scholar 

  5. Van Sant, S.P. & Buckley, P.F. Pharmacotherapy for treatment-refractory schizophrenia. Expert Opin. Pharmacother. 12, 411–434 (2011).

    Article  Google Scholar 

  6. Dong, E., Guidotti, A., Grayson, D.R. & Costa, E. Histone hyperacetylation induces demethylation of reelin and 67-kDa glutamic acid decarboxylase promoters. Proc. Natl. Acad. Sci. USA 104, 4676–4681 (2007).

    Article  CAS  Google Scholar 

  7. Dong, E., Nelson, M., Grayson, D.R., Costa, E. & Guidotti, A. Clozapine and sulpiride but not haloperidol or olanzapine activate brain DNA demethylation. Proc. Natl. Acad. Sci. USA 105, 13614–13619 (2008).

    Article  CAS  Google Scholar 

  8. Abel, T. & Zukin, R.S. Epigenetic targets of HDAC inhibition in neurodegenerative and psychiatric disorders. Curr. Opin. Pharmacol. 8, 57–64 (2008).

    Article  CAS  Google Scholar 

  9. Citrome, L. et al. Adjunctive divalproex and hostility among patients with schizophrenia receiving olanzapine or risperidone. Psychiatr. Serv. 55, 290–294 (2004).

    Article  Google Scholar 

  10. Kelly, D.L. et al. Adjunct divalproex or lithium to clozapine in treatment-resistant schizophrenia. Psychiatr. Q. 77, 81–95 (2006).

    Article  Google Scholar 

  11. Suzuki, T. et al. Augmentation of atypical antipsychotics with valproic acid. An open-label study for most difficult patients with schizophrenia. Hum. Psychopharmacol. 24, 628–638 (2009).

    Article  CAS  Google Scholar 

  12. Löscher, W. Valproate: a reappraisal of its pharmacodynamic properties and mechanisms of action. Prog. Neurobiol. 58, 31–59 (1999).

    Article  Google Scholar 

  13. Gurvich, N., Tsygankova, O.M., Meinkoth, J.L. & Klein, P.S. Histone deacetylase is a target of valproic acid-mediated cellular differentiation. Cancer Res. 64, 1079–1086 (2004).

    Article  CAS  Google Scholar 

  14. Borrelli, E., Nestler, E.J., Allis, C.D. & Sassone-Corsi, P. Decoding the epigenetic language of neuronal plasticity. Neuron 60, 961–974 (2008).

    Article  CAS  Google Scholar 

  15. Arrowsmith, C.H., Bountra, C., Fish, P.V., Lee, K. & Schapira, M. Epigenetic protein families: a new frontier for drug discovery. Nat. Rev. Drug Discov. 11, 384–400 (2012).

    Article  CAS  Google Scholar 

  16. Miyamoto, S., Duncan, G.E., Marx, C.E. & Lieberman, J.A. Treatments for schizophrenia: a critical review of pharmacology and mechanisms of action of antipsychotic drugs. Mol. Psychiatry 10, 79–104 (2005).

    Article  CAS  Google Scholar 

  17. González-Maeso, J. & Sealfon, S.C. Psychedelics and schizophrenia. Trends Neurosci. 32, 225–232 (2009).

    Article  Google Scholar 

  18. Vollenweider, F.X., Vollenweider-Scherpenhuyzen, M.F., Babler, A., Vogel, H. & Hell, D. Psilocybin induces schizophrenia-like psychosis in humans via a serotonin-2 agonist action. Neuroreport 9, 3897–3902 (1998).

    Article  CAS  Google Scholar 

  19. González-Maeso, J. et al. Hallucinogens recruit specific cortical 5-HT(2A) receptor-mediated signaling pathways to affect behavior. Neuron 53, 439–452 (2007).

    Article  Google Scholar 

  20. Moreno, J.L., Sealfon, S.C. & González-Maeso, J. Group II metabotropic glutamate receptors and schizophrenia. Cell. Mol. Life Sci. 66, 3777–3785 (2009).

    Article  CAS  Google Scholar 

  21. Fell, M.J., Svensson, K.A., Johnson, B.G. & Schoepp, D.D. Evidence for the role of metabotropic glutamate (mGlu)2 not mGlu3 receptors in the preclinical antipsychotic pharmacology of the mGlu2/3 receptor agonist (−)-(1R,4S,5S,6S)-4-amino-2-sulfonylbicyclo[3.1.0]hexane-4,6-dicarboxylic acid (LY404039). J. Pharmacol. Exp. Ther. 326, 209–217 (2008).

    Article  CAS  Google Scholar 

  22. Woolley, M.L., Pemberton, D.J., Bate, S., Corti, C. & Jones, D.N. The mGlu2 but not the mGlu3 receptor mediates the actions of the mGluR2/3 agonist, LY379268, in mouse models predictive of antipsychotic activity. Psychopharmacology (Berl.) 196, 431–440 (2008).

    Article  CAS  Google Scholar 

  23. Fribourg, M. et al. Decoding the signaling of a GPCR heteromeric complex reveals a unifying mechanism of action of antipsychotic drugs. Cell 147, 1011–1023 (2011).

    Article  CAS  Google Scholar 

  24. Patil, S.T. et al. Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: a randomized Phase 2 clinical trial. Nat. Med. 13, 1102–1107 (2007).

    Article  CAS  Google Scholar 

  25. González-Maeso, J. et al. Identification of a serotonin/glutamate receptor complex implicated in psychosis. Nature 452, 93–97 (2008).

    Article  Google Scholar 

  26. Agid, O., Kapur, S., Arenovich, T. & Zipursky, R.B. Delayed-onset hypothesis of antipsychotic action: a hypothesis tested and rejected. Arch. Gen. Psychiatry 60, 1228–1235 (2003).

    Article  CAS  Google Scholar 

  27. Egan, M.F. et al. Variation in GRM3 affects cognition, prefrontal glutamate, and risk for schizophrenia. Proc. Natl. Acad. Sci. USA 101, 12604–12609 (2004).

    Article  CAS  Google Scholar 

  28. Liu, W. et al. Pharmacogenetic analysis of the mGlu2/3 agonist LY2140023 monohydrate in the treatment of schizophrenia. Pharmacogenomics J. 12, 246–254 (2012).

    Article  CAS  Google Scholar 

  29. Moreno, J.L. et al. Maternal influenza viral infection causes schizophrenia-like alterations of 5-HT2A and mGlu2 receptors in the adult offspring. J. Neurosci. 31, 1863–1872 (2011).

    Article  CAS  Google Scholar 

  30. Abdolmaleky, H.M. et al. Epigenetic dysregulation of HTR2A in the brain of patients with schizophrenia and bipolar disorder. Schizophr. Res. 129, 183–190 (2011).

    Article  Google Scholar 

  31. Matrisciano, F., Tueting, P., Maccari, S., Nicoletti, F. & Guidotti, A. Pharmacological activation of group-II metabotropic glutamate receptors corrects a schizophrenia-like phenotype induced by prenatal stress in mice. Neuropsychopharmacology 37, 929–938 (2012).

    Article  CAS  Google Scholar 

  32. Huang, H.S., Matevossian, A., Jiang, Y. & Akbarian, S. Chromatin immunoprecipitation in postmortem brain. J. Neurosci. Methods 156, 284–292 (2006).

    Article  CAS  Google Scholar 

  33. Darcy, M.J., Calvin, K., Cavnar, K. & Ouimet, C.C. Regional and subcellular distribution of HDAC4 in mouse brain. J. Comp. Neurol. 518, 722–740 (2010).

    Article  CAS  Google Scholar 

  34. González-Maeso, J. et al. Transcriptome fingerprints distinguish hallucinogenic and nonhallucinogenic 5-hydroxytryptamine 2A receptor agonist effects in mouse somatosensory cortex. J. Neurosci. 23, 8836–8843 (2003).

    Article  Google Scholar 

  35. Tamaru, Y., Nomura, S., Mizuno, N. & Shigemoto, R. Distribution of metabotropic glutamate receptor mGluR3 in the mouse CNS: differential location relative to pre- and postsynaptic sites. Neuroscience 106, 481–503 (2001).

    Article  CAS  Google Scholar 

  36. Benneyworth, M.A. et al. A selective positive allosteric modulator of metabotropic glutamate receptor subtype 2 blocks a hallucinogenic drug model of psychosis. Mol. Pharmacol. 72, 477–484 (2007).

    Article  CAS  Google Scholar 

  37. Aghajanian, G.K. Modeling “psychosis” in vitro by inducing disordered neuronal network activity in cortical brain slices. Psychopharmacology (Berl.) 206, 575–585 (2009).

    Article  CAS  Google Scholar 

  38. Zhai, Y. et al. Group II metabotropic glutamate receptor modulation of DOI-induced c-fos mRNA and excitatory responses in the cerebral cortex. Neuropsychopharmacology 28, 45–52 (2003).

    Article  CAS  Google Scholar 

  39. Kristiansen, L.V., Huerta, I., Beneyto, M. & Meador-Woodruff, J.H. NMDA receptors and schizophrenia. Curr. Opin. Pharmacol. 7, 48–55 (2007).

    Article  CAS  Google Scholar 

  40. Nestler, E.J. & Hyman, S.E. Animal models of neuropsychiatric disorders. Nat. Neurosci. 13, 1161–1169 (2010).

    Article  CAS  Google Scholar 

  41. Geyer, M.A. & Vollenweider, F.X. Serotonin research: contributions to understanding psychoses. Trends Pharmacol. Sci. 29, 445–453 (2008).

    Article  CAS  Google Scholar 

  42. Ludewig, K., Geyer, M.A. & Vollenweider, F.X. Deficits in prepulse inhibition and habituation in never-medicated, first-episode schizophrenia. Biol. Psychiatry 54, 121–128 (2003).

    Article  Google Scholar 

  43. Chiechio, S. et al. Epigenetic modulation of mGlu2 receptors by histone deacetylase inhibitors in the treatment of inflammatory pain. Mol. Pharmacol. 75, 1014–1020 (2009).

    Article  CAS  Google Scholar 

  44. Tsankova, N., Renthal, W., Kumar, A. & Nestler, E.J. Epigenetic regulation in psychiatric disorders. Nat. Rev. Neurosci. 8, 355–367 (2007).

    Article  CAS  Google Scholar 

  45. Guan, J.S. et al. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature 459, 55–60 (2009).

    Article  CAS  Google Scholar 

  46. Gräff, J. et al. An epigenetic blockade of cognitive functions in the neurodegenerating brain. Nature 483, 222–226 (2012).

    Article  Google Scholar 

  47. Peter, C.J. & Akbarian, S. Balancing histone methylation activities in psychiatric disorders. Trends Mol. Med. 17, 372–379 (2011).

    Article  CAS  Google Scholar 

  48. Marder, E. & Taylor, A.L. Multiple models to capture the variability in biological neurons and networks. Nat. Neurosci. 14, 133–138 (2011).

    Article  CAS  Google Scholar 

  49. Dean, B. The cortical serotonin2A receptor and the pathology of schizophrenia: a likely accomplice. J. Neurochem. 85, 1–13 (2003).

    Article  CAS  Google Scholar 

  50. Goldberg, T.E. et al. The effect of clozapine on cognition and psychiatric symptoms in patients with schizophrenia. Br. J. Psychiatry 162, 43–48 (1993).

    Article  CAS  Google Scholar 

  51. Tsankova, N.M. et al. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nat. Neurosci. 9, 519–525 (2006).

    Article  CAS  Google Scholar 

  52. Humphrey, G.W. et al. Complementary roles for histone deacetylases 1, 2, and 3 in differentiation of pluripotent stem cells. Differentiation 76, 348–356 (2008).

    Article  CAS  Google Scholar 

  53. Deacon, R.M. & Rawlins, J.N. T-maze alternation in the rodent. Nat. Protoc. 1, 7–12 (2006).

    Article  Google Scholar 

  54. Hof, P.R., Young, W.G., Bloom, F.E., Belichenko, P.V. & Celio, M.R. Comparative Cytoarchitectonic Atlas of the C57BL/6 and 129/Sv Mouse Brains (Elsevier, Amsterdam, 2000).

  55. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders: DSM-IV 4th edn. (Washington, DC, 1994).

  56. Stan, A.D. et al. Human postmortem tissue: what quality markers matter? Brain Res. 1123, 1–11 (2006).

    Article  CAS  Google Scholar 

  57. Cao, J.L. et al. Mesolimbic dopamine neurons in the brain reward circuit mediate susceptibility to social defeat and antidepressant action. J. Neurosci. 30, 16453–16458 (2010).

    Article  CAS  Google Scholar 

  58. Martin, W.R., Wikler, A., Eades, C.G. & Pescor, F.T. Tolerance to and physical dependence on morphine in rats. Psychopharmacologia 4, 247–260 (1963).

    Article  CAS  Google Scholar 

  59. González-Maeso, J., Rodriguez-Puertas, R., Meana, J.J., Garcia-Sevilla, J.A. & Guimon, J. Neurotransmitter receptor-mediated activation of G-proteins in brains of suicide victims with mood disorders: selective supersensitivity of alpha(2A)-adrenoceptors. Mol. Psychiatry 7, 755–767 (2002).

    Article  Google Scholar 

  60. Shaffer, J.P. Multiple hypothesis testing. Annu. Rev. Psychol. 46, 561–584 (1995).

    Article  Google Scholar 

Download references

Acknowledgements

The authors would like to thank A. Chess, Y. Hurd, C. Alberini, P. Bos and J. Greally for critical review of the manuscript; M. Shapiro for help in working memory experiments; S. Morgello (Mount Sinai School of Medicine) and the Manhattan HIV Brain Bank for providing control brain cortex; P. Casaccia (Mount Sinai School of Medicine) for discussions and for providing the plasmid encoding mouse HDAC2; J. Gingrich (Columbia University) for his gift of 5HT2A−/− mice; V. Rayannavar for assistance with biochemical and behavioral assays; and the staff members of the Basque Institute of Legal Medicine for their cooperation in the study. US National Institutes of Health (NIH) R01 MH084894 (J.G.-M.), Dainippon Sumitomo Pharma (J.G.-M.), NARSAD (J.G.-M.), the Maltz Family Foundation (J.G.-M.), NIH R01 MH090264 (S.J.R.), MICINN SAF2009-084609 (J.J.M.), the Basque Government IT-199-07 (J.J.M.), NIH P50 MH090963 (E.J.N.), NIH R01 MH092306 (M.-H.H.) and NIH P50 MH066392 (J.D.B.) helped fund this study. A.K.F. was supported by NIH grant F32 MH096464. A.G.-B., J.L.M. and G.M. were recipients of predoctoral and postdoctoral fellowships from the Basque government, Ministerio de Ciencia e Innovación, and Consejo Superior de Investigaciones Científicas (CSIC), Spain.

Author information

Authors and Affiliations

Authors

Contributions

M.K. and J.G.-M. designed experiments, analyzed data and wrote the manuscript. M.K. performed experiments. J.G.-M. supervised the research. T.H. performed behavior experiments. A.G.-B. performed experiments in post-mortem schizophrenia-affected brain. A.K. assisted with cloning and performed promoter assay experiments. J.L.M. performed radioligand binding experiments. M.H. assisted with ChIP and DNA methylation assays. G.M., A.M.G., J.H., A.U. and A.L.G. assisted with experiments. R.L.N. performed viral packaging. L.S. performed biostatistical analyses. L.F.C. and J.J.M. obtained and classified post-mortem human brain samples. S.A.G., supervised by S.J.R., helped with stereotaxic surgery. P.J.K. and D.M.D., supervised by E.J.N., helped with viral overexpression. A.K.F., supervised by M.-H.H., performed electrophysiological studies. N.T., supervised by J.D.B., helped with prepulse inhibition test. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Javier González-Maeso.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8, Supplementary Tables 1–2 (PDF 4358 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kurita, M., Holloway, T., García-Bea, A. et al. HDAC2 regulates atypical antipsychotic responses through the modulation of mGlu2 promoter activity. Nat Neurosci 15, 1245–1254 (2012). https://doi.org/10.1038/nn.3181

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3181

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