Generic placeholder image

CNS & Neurological Disorders - Drug Targets

Editor-in-Chief

ISSN (Print): 1871-5273
ISSN (Online): 1996-3181

Review Article

Epigenetic Basis of Psychiatric Disorders: A Narrative Review

Author(s): Fabio Panariello, Giuseppe Fanelli, Chiara Fabbri, Anna Rita Atti, Diana De Ronchi and Alessandro Serretti*

Volume 21, Issue 4, 2022

Published on: 01 December, 2021

Page: [302 - 315] Pages: 14

DOI: 10.2174/1871527320666210825101915

Price: $65

Abstract

Background: Psychiatric disorders are complex, multifactorial illnesses with a demonstrated biological component in their etiopathogenesis. Epigenetic modifications, through the modulation of DNA methylation, histone modifications and RNA interference, tune tissue-specific gene expression patterns and play a relevant role in the etiology of psychiatric illnesses.

Objective: This review aims to discuss the epigenetic mechanisms involved in psychiatric disorders, their modulation by environmental factors and their interactions with genetic variants, in order to provide a comprehensive picture of their mutual crosstalk.

Methods: In accordance with the PRISMA guidelines, systematic searches of Medline, EMBASE, PsycINFO, Web of Science, Scopus, and the Cochrane Library were conducted.

Results: Exposure to environmental factors, such as poor socio-economic status, obstetric complications, migration, and early life stressors, may lead to stable changes in gene expression and neural circuit function, playing a role in the risk of psychiatric diseases. The most replicated genes involved by studies using different techniques are discussed. Increasing evidence indicates that these sustained abnormalities are maintained by epigenetic modifications in specific brain regions and they interact with genetic variants in determining the risk of psychiatric disorders.

Conclusion: An increasing amount of evidence suggests that epigenetics plays a pivotal role in the etiopathogenesis of psychiatric disorders. New therapeutic approaches may work by reversing detrimental epigenetic changes that occurred during the lifespan.

Keywords: Epigenetics, methylation, schizophrenia, mood disorders, stress, psychiatric disorders.

Graphical Abstract
[1]
Karlsgodt KH, Sun D, Cannon TD. Structural and functional brain abnormalities in schizophrenia. Curr Dir Psychol Sci 2010; 19: 226-31.
[2]
Rehm J, Kevin D. Shield. global burden of disease and the impact of mental and addictive disorders. Curr Psychiatry Rep 2019; 21(2): 10.
[3]
Papadimitriou G. The ‘Biopsychosocial Model’: 40 years of application in Psychiatry. Psychiatr Psychiatr 2017; 28: 107-10.
[4]
Peña CJ, Bagot RC, Labonté B, Nestler EJ. Epigenetic signaling in psychiatric disorders. J Mol Biol 2014; 426: 3389-412.
[5]
Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 2003; 33(Suppl.): 245-54.
[6]
Jenuwein T, Allis CD. Translating the histone code. Science 2001; 293: 1074-80.
[7]
Kiser DP, Rivero O, Lesch K-P. Annual research review: The (epi)genetics of neurodevelopmental disorders in the era of whole-genome sequencing-unveiling the dark matter. J Child Psychol Psychiatry 2015; 56: 278-95.
[8]
Moher D, Liberati A, Tetzlaff J, Altman DG. PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med 2009; 6: e1000097.
[9]
Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 2007; 447: 425-32.
[10]
Suzuki MM, Bird A. DNA methylation landscapes: provocative insights from epigenomics. Nat Rev Genet 2008; 9: 465-76.
[11]
Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 2012; 13: 484-92.
[12]
Neri F, Rapelli S, Krepelova A, Incarnato D, Parlato C, Basile G, et al. Intragenic DNA methylation prevents spurious transcription initiation. Nature 2017; 543: 72-7.
[13]
Maunakea AK, Chepelev I, Cui K, Zhao K. Intragenic DNA methylation modulates alternative splicing by recruiting MeCP2 to promote exon recognition. Cell Res 2013; 23: 1256-69.
[14]
Klengel T, Binder EB. Epigenetics of stress-related psychiatric disorders and gene × environment interactions. Neuron 2015; 86: 1343-57.
[15]
Guidotti A, Grayson DR, Caruncho HJ. Epigenetic RELN dysfunction in schizophrenia and related neuropsychiatric disorders. Front Cell Neurosci 2016; 10: 89.
[16]
Costa E, Chen Y, Davis J, et al. REELIN and schizophrenia: A disease at the interface of the genome and the epigenome. Mol Interv 2002; 2: 47-57.
[17]
Grayson DR, Chen Y, Costa E, et al. The human reelin gene: Transcription factors (+), repressors (-) and the methylation switch (+/-) in schizophrenia. Pharmacol Ther 2006; 111: 272-86.
[18]
Feng J, Zhou Y, Campbell SL, et al. Dnmt1 and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons. Nat Neurosci 2010; 13: 423-30.
[19]
Kundakovic M, Chen Y, Costa E, Grayson DR. DNA methyltransferase inhibitors coordinately induce expression of the human reelin and glutamic acid decarboxylase 67 genes. Mol Pharmacol 2007; 71: 644-53.
[20]
Huang H-S, Akbarian S. GAD1 mRNA expression and DNA methylation in prefrontal cortex of subjects with schizophrenia. PLoS One 2007; 2(8): e809.
[21]
Pries L-K, Gülöksüz S, Kenis G. DNA Methylation in Schizophrenia. Adv Exp Med Biol 2017; 978: 211-36.
[22]
Petronis A, Gottesman II, Kan P, et al. Monozygotic twins exhibit numerous epigenetic differences: clues to twin discordance? Schizophr Bull 2003; 29: 169-78.
[23]
Abdolmaleky HM, Smith CL, Zhou J-R, Thiagalingam S. Epigenetic alterations of the dopaminergic system in major psychiatric disorders. Methods Mol Biol 2008; 448: 187-212.
[24]
Pai S, Li P, Killinger B, et al. Differential methylation of enhancer at IGF2 is associated with abnormal dopamine synthesis in major psychosis. Nat Commun 2019; 10: 2046.
[25]
Abdolmaleky HM, Yaqubi S, Papageorgis P, et al. Epigenetic dysregulation of HTR2A in the brain of patients with schizophrenia and bipolar disorder. Schizophr Res 2011; 129: 183-90.
[26]
Iwamoto K, Bundo M, Yamada K, et al. A family-based and case- control association study of SOX10 in schizophrenia. Am J Med Genet Part B Neuropsychiatr Genet Off Publ Int Soc Psychiatr Genet 2006; 141B: 477-81.
[27]
Richardson TG, Shihab HA, Hemani G, et al. Collapsed methylation quantitative trait loci analysis for low frequency and rare variants. Hum Mol Genet 2016; 25: 4339-49.
[28]
Mendizabal I, Berto S, Usui N, et al. Cell type-specific epigenetic links to schizophrenia risk in the brain. Genome Biol 2019; 20: 135.
[29]
Jaffe AE, Gao Y, Deep-Soboslay A, et al. Mapping DNA methylation across development, genotype and schizophrenia in the human frontal cortex. Nat Neurosci 2016; 19: 40-7.
[30]
Khavari B, Cairns MJ. Epigenomic dysregulation in schizophrenia: in search of disease etiology and biomarkers. Cells 2020; 9(8): 1837.
[31]
Wockner LF, Noble EP, Lawford BR, et al. Genome-wide DNA methylation analysis of human brain tissue from schizophrenia patients. Transl Psychiatry 2014; 4(1): e339.
[32]
Pidsley R, Viana J, Hannon E, et al. Methylomic profiling of human brain tissue supports a neurodevelopmental origin for schizophrenia. Genome Biol 2014; 15: 483.
[33]
Hannon E, Dempster E, Viana J, et al. An integrated genetic-epigenetic analysis of schizophrenia: evidence for co-localization of genetic associations and differential DNA methylation. Genome Biol 2016; 17: 176.
[34]
Aberg KA, McClay JL, Nerella S, et al. Methylome-wide association study of schizophrenia: identifying blood biomarker signatures of environmental insults. JAMA Psychiatry 2014; 71: 255-64.
[35]
Kinoshita M, Numata S, Tajima A, et al. Aberrant DNA methylation of blood in schizophrenia by adjusting for estimated cellular proportions. Neuromolecular Med 2014; 16: 697-703.
[36]
Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat Methods 2013; 10: 1213-8.
[37]
Bryois J, Garrett ME, Song L, et al. Evaluation of chromatin accessibility in prefrontal cortex of individuals with schizophrenia. Nat Commun 2018; 9: 3121.
[38]
Park C, Rosenblat JD, Brietzke E, et al. Stress, epigenetics and depression: A systematic review. Neurosci Biobehav Rev 2019; 102: 139-52.
[39]
McGowan PO, Sasaki A, D’Alessio AC, et al. Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat Neurosci 2009; 12: 342-8.
[40]
Li M, D’Arcy C, Li X, Zhang T, Joober R, Meng X. What do DNA methylation studies tell us about depression? A systematic review. Transl Psychiatry 2019; 9: 68.
[41]
Sugawara H, Iwamoto K, Bundo M, et al. Hypermethylation of serotonin transporter gene in bipolar disorder detected by epigenome analysis of discordant monozygotic twins. Transl Psychiatry 2011; 1(7): e24.
[42]
Egervari G, Kozlenkov A, Dracheva S, Hurd YL. Molecular windows into the human brain for psychiatric disorders. Mol Psychiatry 2019; 24: 653-73.
[43]
Aberg KA, Shabalin AA, Chan RF, et al. Convergence of evidence from a methylome-wide CpG-SNP association study and GWAS of major depressive disorder. Transl Psychiatry 2018; 8: 162.
[44]
Gore BB, Miller SM, Jo YS, et al. Roundabout receptor 2 maintains inhibitory control of the adult midbrain. eLife 2017; 6: e23858.
[45]
Meda SA, Ruaño G, Windemuth A, et al. Multivariate analysis reveals genetic associations of the resting default mode network in psychotic bipolar disorder and schizophrenia. Proc Natl Acad Sci USA 2014; 111: E2066-207.
[46]
Torres-Berrío A, Lopez JP, Bagot RC, et al. DCC Confers Susceptibility to Depression-like Behaviors in Humans and Mice and Is Regulated by miR-218. Biol Psychiatry 2017; 81(4): 306-15.
[47]
Sharma RP, Rosen C, Kartan S, et al. Valproic acid and chromatin remodeling in schizophrenia and bipolar disorder: preliminary results from a clinical population. Schizophr Res 2006; 88: 227-31.
[48]
Wang Y, Xia J, Helfer B, Li C, Leucht S. Valproate for schizophrenia. Cochrane Database Syst Rev 2016; 11: CD004028.
[49]
Gilbert TM, Zürcher NR, Wu CJ, Bhanot A, Hightower BG, Kim M. PET neuroimaging reveals histone deacetylase dysregulation in schizophrenia. J Clin Invest 2019; 129(1): 364-72.
[50]
Akbarian S, Ruehl MG, Bliven E, et al. Chromatin alterations associated with down-regulated metabolic gene expression in the prefrontal cortex of subjects with schizophrenia. Arch Gen Psychiatry 2005; 62: 829-40.
[51]
Sharma RP, Grayson DR, Gavin DP. Histone deactylase 1 expression is increased in the prefrontal cortex of schizophrenia subjects: analysis of the National Brain Databank microarray collection. J Psychiatr Res 2015; 65: 87-94.
[52]
Chase KA, Rosen C, Rubin LH, et al. Evidence of a sex-dependent restrictive epigenome in schizophrenia. J Psychiatr Res 2015; 65: 87-94.
[http://dx.doi.org/10.1016/j.jpsychires.2015.04.005]
[53]
Girdhar K, Hoffman GE, Jiang Y, et al. Cell-specific histone modification maps in the human frontal lobe link schizophrenia risk to the neuronal epigenome. Nat Neurosci 2018; 21: 1126-36.
[54]
Chase KA, Gavin DP, Guidotti A, Sharma RP. Histone methylation at H3K9: evidence for a restrictive epigenome in schizophrenia. Schizophr Res 2013; 149(1-3): 15-20.
[http://dx.doi.org/10.1016/j.schres.2013.06.021]
[55]
Covington HE, Vialou VF, LaPlant Q, Ohnishi YN, Nestler EJ. Hippocampal-dependent antidepressant-like activity of histone deacetylase inhibition. Neurosci Lett 2011; 493: 122-6.
[56]
Robison AJ, Vialou V, Sun H-S, et al. Fluoxetine epigenetically alters the CaMKIIα promoter in nucleus accumbens to regulate ΔFosB binding and antidepressant effects. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol 2014; 39: 1178-86.
[57]
Cruceanu C, Freemantle E, Alda M, Rouleau GA, Turecki G. Epigenetic regulation of synapsin genes in mood disorders. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol 2013; 38: 239-41.
[58]
Cech TR, Steitz JA. The noncoding RNA revolution-trashing old rules to forge new ones. Cell 2014; 157: 77-94.
[59]
Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004; 116: 281-97.
[60]
Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005; 120: 15-20.
[61]
Lim LP, Lau NC, Garrett-Engele P, et al. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 2005; 433: 769-73.
[62]
Kapranov P, Ozsolak F, Milos PM. Profiling of short RNAs using Helicos single-molecule sequencing. Methods Mol Biol 2012; 822: 219-32.
[63]
Plath K, Fang J, Mlynarczyk-Evans SK, et al. Role of histone H3 lysine 27 methylation in X inactivation. Science 2003; 300: 131-5.
[64]
de Napoles M, Mermoud JE, Wakao R, et al. Polycomb group proteins Ring1A/B link ubiquitylation of histone H2A to heritable gene silencing and X inactivation. Dev Cell 2004; 7: 663-76.
[65]
Rinn JL, Kertesz M, Wang JK, et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 2007; 129: 1311-23.
[66]
Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet 2009; 10: 155-9.
[67]
Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell 2009; 136: 629-41.
[68]
Smalheiser NR. The RNA-centred view of the synapse: non-coding RNAs and synaptic plasticity. Philos Trans R Soc Lond B Biol Sci 2014; 369(1652): 20130504.
[69]
Issler O, Chen A. Determining the role of microRNAs in psychiatric disorders. Nat Rev Neurosci 2015; 16: 201-12.
[70]
Sheinerman KS, Umansky SR. Circulating cell-free microRNA as biomarkers for screening, diagnosis and monitoring of neurodegenerative diseases and other neurologic pathologies. Front Cell Neurosci 2013; 7: 150.
[71]
Maffioletti E, Cattaneo A, Rosso G, et al. Peripheral whole blood microRNA alterations in major depression and bipolar disorder. J Affect Disord 2016; 200: 250-8.
[72]
Sun X, Lu J, Zhang L, et al. Aberrant microRNA expression in peripheral plasma and mononuclear cells as specific blood-based biomarkers in schizophrenia patients. J Clin Neurosci Off J Neurosurg Soc Australas 2015; 22: 570-4.
[73]
Burmistrova OA, Goltsov AY, Abramova LI, Kaleda VG, Orlova VA, Rogaev EI. MicroRNA in schizophrenia: genetic and expression analysis of miR-130b (22q11). Biochem Biokhimiia 2007; 72: 578-82.
[74]
Van L, Boot E, Bassett AS. Update on the 22q11.2 deletion syndrome and its relevance to schizophrenia. Curr Opin Psychiatry 2017; 30: 191-6.
[75]
Beveridge NJ, Tooney PA, Carroll AP, et al. Dysregulation of miRNA 181b in the temporal cortex in schizophrenia. Hum Mol Genet 2008; 17: 1156-68.
[76]
Banigan MG, Kao PF, Kozubek JA, et al. Differential expression of exosomal microRNAs in prefrontal cortices of schizophrenia and bipolar disorder patients. PLoS One 2013; 8(1): e48814.
[77]
Forstner AJ, Degenhardt F, Schratt G, Nöthen MM. MicroRNAs as the cause of schizophrenia in 22q11.2 deletion carriers, and possible implications for idiopathic disease: a mini-review. Front Mol Neurosci 2013; 6: 47.
[78]
Topol A, Zhu S, Hartley BJ, et al. Dysregulation of miRNA-9 in a Subset of Schizophrenia Patient-Derived Neural Progenitor Cells. Cell Rep 2017; 20: 2525.
[79]
Alacam H, Akgun S, Akca H, Ozturk O, Kabukcu BB, Herken H. miR-181b-5p, miR-195-5p and miR-301a-3p are related with treatment resistance in schizophrenia. Psychiatry Res 2016; 245: 200-6.
[80]
Liu Y, Chang X, Hahn C-G, Gur RE, Sleiman PAM, Hakonarson H. Non-coding RNA dysregulation in the amygdala region of schizophrenia patients contributes to the pathogenesis of the disease. Transl Psychiatry 2018; 8: 44.
[81]
Sakamoto K, Crowley JJ. A comprehensive review of the genetic and biological evidence supports a role for MicroRNA-137 in the etiology of schizophrenia. Am J Med Genet Part B Neuropsychiatr Genet Off Publ Int Soc Psychiatr Genet 2018; 177: 242-56.
[82]
Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014; 511(7510): 421-7.
[83]
Wright BJ, Jackson MB. Voltage imaging in the study of hippocampal circuit function and plasticity. Adv Exp Med Biol 2015; 859: 197-211.
[84]
Landek-Salgado MA, Faust TE, Sawa A. Molecular substrates of schizophrenia: homeostatic signaling to connectivity. Mol Psychiatry 2016; 21: 10-28.
[85]
Du K, Zhao C, Wang L, et al. MiR-191 inhibit angiogenesis after acute ischemic stroke targeting VEZF1. Aging (Albany NY) 2019; 11: 2762-86.
[86]
Tsuang MT, Faraone SV. The case for heterogeneity in the etiology of schizophrenia. Schizophr Res 1995; 17: 161-75.
[87]
Kaur G, Begum R, Thota S, Batra S. A systematic review of smoking-related epigenetic alterations. Arch Toxicol 2019; 93: 2715-40.
[88]
Sun E, Shi Y. MicroRNAs: Small molecules with big roles in neurodevelopment and diseases. Exp Neurol 2015; 268: 46-53.
[89]
Fang Y, Qiu Q, Zhang S, et al. Changes in miRNA-132 and miR-124 levels in non-treated and citalopram-treated patients with depression. J Affect Disord 2018; 227: 745-51.
[90]
Qi S, Yang X, Zhao L, et al. MicroRNA132 associated multimodal neuroimaging patterns in unmedicated major depressive disorder. Brain 2018; 141: 916-26.
[91]
Molendijk ML, Spinhoven P, Polak M. Bus B a. A, Penninx BWJH, Elzinga BM. Serum BDNF concentrations as peripheral manifestations of depression: evidence from a systematic review and meta-analyses on 179 associations (N=9484). Mol Psychiatry 2014; 19: 791-800.
[92]
Azevedo JA, Carter BS, Meng F, et al. The microRNA network is altered in anterior cingulate cortex of patients with unipolar and bipolar depression. J Psychiatr Res 2016; 82: 58-67.
[93]
Fiori LM, Lopez JP, Richard-Devantoy S, et al. Investigation of miR-1202, miR-135a, and miR-16 in Major Depressive Disorder and Antidepressant Response. Int J Neuropsychopharmacol 2017; 20: 619-23.
[94]
Kendler KS. Hierarchy and heritability: the role of diagnosis and modeling in psychiatric genetics. Am J Psychiatry 2002; 159: 515-8.
[95]
Belsky J, Steinberg LD, Houts RM, et al. Family rearing antecedents of pubertal timing. Child Dev 2007; 78: 1302-21.
[96]
[97]
Richetto J, Meyer U. Epigenetic Modifications in Schizophrenia and Related Disorders: Molecular Scars of Environmental Exposures and Source of Phenotypic Variability. Biol Psychiatry 2021; 89: 215-26.
[98]
Vineis P, Chatziioannou A, Cunliffe VT, et al. Epigenetic memory in response to environmental stressors. FASEB J 2017; 31: 2241-51.
[99]
de Kluiver H, Buizer-Voskamp JE, Dolan CV, Boomsma DI. Paternal age and psychiatric disorders: A review. Am J Med Genet Part B Neuropsychiatr Genet Off Publ Int Soc Psychiatr Genet 2017; 174: 202-13.
[100]
Assary E, Vincent JP, Keers R, Pluess M. Gene-environment interaction and psychiatric disorders: Review and future directions. Semin Cell Dev Biol 2018; 77: 133-43.
[101]
Carr CP, Martins CMS, Stingel AM, Lemgruber VB, Juruena MF. The role of early life stress in adult psychiatric disorders: a systematic review according to childhood trauma subtypes. J Nerv Ment Dis 2013; 201: 1007-20.
[102]
Perrin JM, Homer CJ. The quality of children’s health care matters-time to pay attention. N Engl J Med 2007; 357: 1549-51.
[103]
Gerra MC, Jayanthi S, Manfredini M, et al. Gene variants and educational attainment in cannabis use: mediating role of DNA methylation. Transl Psychiatry 2018; 8: 23.
[104]
Razin A, Riggs AD. DNA methylation and gene function. Science 1980; 210: 604-10.
[105]
Strahl BD, Allis CD. The language of covalent histone modifications. Nature 2000; 403: 41-5.
[106]
Tsankova NM, Berton O, Renthal W, Kumar A, Neve RL, Nestler EJ. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nat Neurosci 2006; 9: 519-25.
[107]
Klengel T, Mehta D, Anacker C, et al. Allele-specific FKBP5 DNA demethylation mediates gene-childhood trauma interactions. Nat Neurosci 2013; 16: 33-41.
[108]
Klinger-König J, Hertel J, Van der Auwera S, et al. Methylation of the FKBP5 gene in association with FKBP5 genotypes, childhood maltreatment and depression. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol 2019; 44: 930-8.
[109]
Yeo S, Enoch M-A, Gorodetsky E, Akhtar L, Schuebel K, Roy A, et al. The influence of FKBP5 genotype on expression of FKBP5 and other glucocorticoid-regulated genes, dependent on trauma exposure. Genes Brain Behav 2017; 16: 223-32.
[110]
Wang Q, Shelton RC, Dwivedi Y. Interaction between early-life stress and FKBP5 gene variants in major depressive disorder and post-traumatic stress disorder: A systematic review and meta-analysis. J Affect Disord 2018; 225: 422-8.
[111]
Mumtaz F, Khan MI, Zubair M, Dehpour AR. Neurobiology and consequences of social isolation stress in animal model-A comprehensive review. Biomed Pharmacother Biomedecine Pharmacother 2018; 105: 1205-22.
[112]
Sullivan PF, Geschwind DH. Defining the genetic, genomic, cellular, and diagnostic architectures of psychiatric disorders. Cell 2019; 177: 162-83.
[113]
Saradalekshmi KR, Neetha NV, Sathyan S, Nair IV, Nair CM, Banerjee M. DNA methyl transferase (DNMT) gene polymorphisms could be a primary event in epigenetic susceptibility to schizophrenia. PLoS One 2014; 9: e98182.
[114]
Li CH, Coffey EL, Dall’Agnese A, et al. MeCP2 links heterochromatin condensates and neurodevelopmental disease. Nature 2020; 586: 440-4.
[115]
Zhang F, Lupski JR. Non-coding genetic variants in human disease. Hum Mol Genet 2015; 24: R102-10.
[116]
Hancock DB, Guo Y, Reginsson GW, et al. Genome-wide association study across European and African American ancestries identifies a SNP in DNMT3B contributing to nicotine dependence. Mol Psychiatry 2018; 23: 1911-9.
[117]
Swets M, Middeldorp CM, Schoevers RA. [Heritability and environment in psychiatric disorders]. Tijdschr Psychiatr 2009; 51: 651-63.
[118]
Alam R, Abdolmaleky HM, Zhou J-R. Microbiome, inflammation, epigenetic alterations, and mental diseases. Am J Med Genet Part B Neuropsychiatr Genet Off Publ Int Soc Psychiatr Genet 2017; 174: 651-60.
[119]
Huan T, Mendelson M, Joehanes R, et al. Epigenome-wide association study of DNA methylation and microRNA expression highlights novel pathways for human complex traits. Epigenetics 2020; 15: 183-98.
[120]
Harb-de la Rosa A, Acker M, Kumar RA, Manoharan M. Epigenetics application in the diagnosis and treatment of bladder cancer. Can J Urol 2015; 22: 7947-51.
[121]
Szyf M. Prospects for the development of epigenetic drugs for CNS conditions. Nat Rev Drug Discov 2015; 14: 461-74.
[122]
Majumder A, Dhara SK, Swetenburg R, et al. Inhibition of DNA methyltransferases and histone deacetylases induces astrocytic differentiation of neural progenitors. Stem Cell Res (Amst) 2013; 11: 574-86.
[123]
Guan J-S, Haggarty SJ, Giacometti E, et al. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature 2009; 459: 55-60.
[124]
Ishimaru N, Fukuchi M, Hirai A, et al. Differential epigenetic regulation of BDNF and NT-3 genes by trichostatin A and 5-aza-2′-deoxycytidine in Neuro-2a cells. Biochem Biophys Res Commun 2010; 394: 173-7.
[125]
Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan TG, Cryan JF. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem Int 2016; 99: 110-32.
[126]
Hantsoo L, Jašarević E, Criniti S, et al. Childhood adversity impact on gut microbiota and inflammatory response to stress during pregnancy. Brain Behav Immun 2019; 75: 240-50.
[127]
Weïwer M, Lewis MC, Wagner FF, Holson EB. Therapeutic potential of isoform selective HDAC inhibitors for the treatment of schizophrenia. Future Med Chem 2013; 5: 1491-508.
[128]
Heerboth S, Lapinska K, Snyder N, Leary M, Rollinson S, Sarkar S. Use of epigenetic drugs in disease: an overview. Genet Epigenet 2014; 6: 9-19.
[129]
Daniels TE, Olsen EM, Tyrka AR. Stress and psychiatric disorders: the role of mitochondria. Annu Rev Clin Psychol 2020; 16: 165-86.
[130]
Picard M, McEwen BS. Psychological stress and mitochondria: a conceptual framework. BMC Psychosom Med 2018; 80(2): 126-40.
[131]
Tolosa A, Sanjuán J, Dagnall AM, Moltó MD, Herrero N, de Frutos R. FOXP2 gene and language impairment in schizophrenia: Association and epigenetic studies. BMC Med Genet 2010; 11: 114.
[132]
Sanjuán J, Castro-Martínez XH, García-Martí G, et al. FOXP2 expression and gray matter density in the male brains of patients with schizophrenia. Brain Imaging Behav 2020; 2020: 30.
[http://dx.doi.org/10.1007/s11682-020-00339-x]
[133]
Mill J, Tang T, Kaminsky Z, et al. Epigenomic profiling reveals DNA-methylation changes associated with major psychosis. Am J Hum Genet 2008; 82: 696-711.
[134]
Gürel Ç, Kuşçu GC, Yavaşoğlu A, Biray Avcı Ç. The clues in solving the mystery of major psychosis: The epigenetic basis of schizophrenia and bipolar disorder. Neurosci Biobehav Rev 2020; 113: 51-61.
[135]
Rao JS, Keleshian VL, Klein S, Rapoport SI. Epigenetic modifications in frontal cortex from Alzheimer’s disease and bipolar disorder patients. Transl Psychiatry 2012; 2(7): e132.
[136]
Ghadirivasfi M, Nohesara S, Ahmadkhaniha H-R, et al. Hypomethylation of the serotonin receptor type-2A Gene (HTR2A) at T102C polymorphic site in DNA derived from the saliva of patients with schizophrenia and bipolar disorder. Am J Med Genet Part B Neuropsychiatr Genet Off Publ Int Soc Psychiatr Genet 2011; 156B: 536-45.
[137]
Kaminsky Z, Jones I, Verma R, et al. DNA methylation and expression of KCNQ3 in bipolar disorder. Bipolar Disord 2015; 17: 150-9.
[138]
Ludwig B, Dwivedi Y. Dissecting bipolar disorder complexity through epigenomic approach. Mol Psychiatry 2016; 21: 1490-8.
[139]
Popendikyte V, Laurinavicius A, Paterson AD, Macciardi F, Kennedy JL, Petronis A. DNA methylation at the putative promoter region of the human dopamine D2 receptor gene. Neuroreport 1999; 10: 1249-55.
[140]
Nohesara S, Ghadirivasfi M, Mostafavi S, et al. DNA hypomethylation of MB- COMT promoter in the DNA derived from saliva in schizophrenia and bipolar disorder. J Psychiatr Res 2011; 45: 1432-8.
[141]
Abdolmaleky HM, Cheng K-H, Faraone SV, et al. Hypomethylation of MB-COMT promoter is a major risk factor for schizophrenia and bipolar disorder. Hum Mol Genet 2006; 15: 3132-45.
[142]
Veldic M, Caruncho HJ, Liu WS, et al. DNA-methyltransferase 1 mRNA is selectively overexpressed in telencephalic GABAergic interneurons of schizophrenia brains. Proc Natl Acad Sci USA 2004; 101: 348-53.
[143]
Pal M, Ebrahimi S, Oh G, et al. High Precision DNA Modification Analysis of HCG9 in Major Psychosis. Schizophr Bull 2016; 42: 170-7.
[144]
Voisey J, Lawford B, Bruenig D, et al. Differential BDNF methylation in combat exposed veterans and the association with exercise. Gene 2019; 698: 107-12.
[145]
Starnawska A, Demontis D, McQuillin A, et al. Hypomethylation of FAM63B in bipolar disorder patients. Clin Epigenetics 2016; 8: 52.
[146]
Gao J, Yi H, Tang X, et al. DNA methylation and gene expression of matrix metalloproteinase 9 gene in deficit and non-deficit schizophrenia. Front Genet 2018; 9: 646.
[147]
Fachim HA, Loureiro CM, Corsi-Zuelli F, et al. GRIN2B promoter methylation deficits in early-onset schizophrenia and its association with cognitive function. Epigenomics 2019; 11: 401-10.
[148]
Garcia-Ruiz B, Moreno L, Muntané G, et al. Leukocyte and brain DDR1 hypermethylation is altered in psychosis and is correlated with stress and inflammatory markers. Epigenomics 2020; 12: 251-65.
[149]
Zannas AS, Wiechmann T, Gassen NC, Binder EB. Gene-stress-epigenetic regulation of FKBP5: clinical and translational implications. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol 2016; 41: 261-74.
[150]
Wheater ENW, Stoye DQ, Cox SR, et al. DNA methylation and brain structure and function across the life course: A systematic review. Neurosci Biobehav Rev 2020; 113: 133-56.
[151]
Sadeh N, Spielberg JM, Logue MW, et al. SKA2 methylation predicts reduced cortical thickness in prefrontal cortex. Mol Psychiatry 2016; 21: 299.
[152]
Schechter DS, Moser DA, Pointet VC, et al. The association of serotonin receptor 3A methylation with maternal violence exposure, neural activity, and child aggression. Behav Brain Res 2017; 325: 268-77.
[153]
Yehuda R, Hoge CW, McFarlane AC, et al. Post-traumatic stress disorder. Nat Rev Dis Primers 2015; 1: 15057.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy