We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Dysregulation of schizophrenia-associated genes and genome-wide hypomethylation in neurons overexpressing DNMT1

    Sonal Saxena

    Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, India

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Sumana Choudhury

    Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, India

    Centre for Human Disease Research, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, India

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Pranay Amruth Maroju

    Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, India

    ,
    Anuhya Anne

    Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, India

    Centre for Human Disease Research, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, India

    ,
    Lov Kumar

    Computer Science & Information Systems, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, India

    &
    Kommu Naga Mohan

    *Author for correspondence:

    E-mail Address: kommumohan@gmail.com

    Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, India

    Centre for Human Disease Research, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, India

    Published Online:https://doi.org/10.2217/epi-2021-0133

    Aim: To study the effects of DNMT1 overexpression on transcript levels of genes dysregulated in schizophrenia and on genome-wide methylation patterns. Materials & methods: Transcriptome and DNA methylome comparisons were made between R1 (wild-type) and Dnmt1tet/tet mouse embryonic stem cells and neurons overexpressing DNMT1. Genes dysregulated in both Dnmt1tet/tet cells and schizophrenia patients were studied further. Results & conclusions: About 50% of dysregulated genes in patients also showed altered transcript levels in Tet/Tet neurons in a DNA methylation-independent manner. These neurons unexpectedly showed genome-wide hypomethylation, increased transcript levels of Tet1 and Apobec 1-3 genes and increased activity and copy number of LINE-1 elements. The observed similarities between Tet/Tet neurons and schizophrenia brain samples reinforce DNMT1 overexpression as a risk factor.

    Lay abstract

    DNMT1 controls cytosine methylation, which is often associated with reduced gene expression. Increased levels of DNMT1 is a risk factor for schizophrenia but information on the affected genes is limited. In this study, ∼50% of genes with altered levels of messenger RNAs in schizophrenia patients were also altered in neurons with increased DNMT1. Surprisingly, the neurons with higher DNMT1 levels showed genome-wide decrease in methylation. These findings uncover a new type of gene dysregulation that is independent of DNMT1's catalytic activity.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Murray CJ, Lopez AD. A comprehensive assessment of mortality and disability from diseases, injuries, and risk factors in 1990 and projected to 2020. In: Global Burden of Disease and Injury Series. Harvard Unversity Press, ISBN 0-9655466-0-8, 1–43 (1996).
    • 2. Cardno AG, Gottesman II. Twin studies of schizophrenia: from bow-and-arrow concordances to star wars Mx and functional genomics. Am. J. Med. Genet. 97(1), 12–17 (2000).
    • 3. Zhang Y, Reinberg D. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev. 15(18), 2343–2360 (2001).
    • 4. Bestor TH, Edwards JR, Boulard M. Notes on the role of dynamic DNA methylation in mammalian development. Proc. Natl Acad. Sci. USA 112(22), 6796–6799 (2015).
    • 5. Seto E, Yoshida M. Erasers of histone acetylation: the histone deacetylase enzymes. Cold Spring Harb. Perspect. Biol. 6(4), a018713 (2014).
    • 6. Maltby VE, Martin BJE, Brind'Amour J et al. Histone H3K4 demethylation is negatively regulated by histone H3 acetylation in Saccharomyces cerevisiae. Proc. Natl Acad. Sci. USA 109(45), 18505–18510 (2012).
    • 7. Schomacher L. Mammalian DNA demethylation: multiple faces and upstream regulation. Epigenetics 8(7), 679–684 (2013).
    • 8. Zhang L, Lu Q, Chang C. Epigenetics in health and disease. Adv. Exp. Med. Biol. 1253, 3–55 (2020).
    • 9. Torabi Moghadam B, Etemadikhah M, Rajkowska G et al. Analyzing DNA methylation patterns in subjects diagnosed with schizophrenia using machine learning methods. J. Psychiatr. Res. 114, 41–47 (2019).
    • 10. 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 62(8), 829–840 (2005).
    • 11. Perkins DO, Jeffries CD, Jarskog LF et al. microRNA expression in the prefrontal cortex of individuals with schizophrenia and schizoaffective disorder. Genome Biol. 8(2), R27 (2007).
    • 12. Bahari-Javan S, Varbanov H, Halder R et al. HDAC1 links early life stress to schizophrenialike phenotypes. Proc. Natl Acad. Sci. USA 114(23), E4686–E4694 (2017).
    • 13. Schroeder FA, Gilbert TM, Feng N et al. Expression of HDAC2 but not HDAC1 transcript is reduced in dorsolateral prefrontal cortex of patients with schizophrenia. ACS Chem. Neurosci. 8(3), 662–668 (2017).
    • 14. 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. Schizophr. Res. 98(1–3), 111–117 (2008).
    • 15. Zhubi A, Veldic M, Puri NV et al. An upregulation of DNA-methyltransferase 1 and 3a expressed in telencephalic GABAergic neurons of schizophrenia patients is also detected in peripheral blood lymphocytes. Schizophr. Res. 111(1–3), 115–122 (2009).
    • 16. 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 101(1), 348–353 (2004).
    • 17. Veldic M, Guidotti A, Maloku E, Davis JM, Costa E. In psychosis, cortical interneurons overexpress DNA-methyltransferase 1. Proc. Natl Acad. Sci. USA 102(6), 2152–2157 (2005). •• This was one of the first studies reporting overexpression of DNMT1 in the postmortem brain samples of schizophrenia patients.
    • 18. 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. 71(3), 644–653 (2007). • This study provides evidence in support of the use of DNMT inhibitors in treating psychosis.
    • 19. Biniszkiewicz D, Gribnau J, Ramsahoye B et al. DNMT1 overexpression causes genomic hypermethylation, loss of imprinting, and embryonic lethality. Mol. Cell. Biol. 22(7), 2124–2135 (2002).
    • 20. D'Aiuto L, Di Maio R, Mohan KN et al. Mouse ES cells overexpressing DNMT1 produce abnormal neurons with upregulated NMDA/NR1 subunit. Differentiation 82(1), 9–17 (2011). •• This is the first report that provides evidence in support of DNMT1 overexpression and abnormal neurogenesis.
    • 21. Sen GL, Reuter JA, Webster DE, Zhu L, Khavari PA. DNMT1 maintains progenitor function in self-renewing somatic tissue. Nature 463(7280), 563–567 (2010).
    • 22. Mohan KN. Stem cell models to investigate the role of DNA methylation machinery in development of neuropsychiatric disorders. Stem Cells Int. 2016, 4379425 (2016).
    • 23. Saxena S, Maroju PA, Choudhury S, Anne A, Mohan KN. Analysis of transcript levels of a few schizophrenia candidate genes in neurons from a transgenic mouse embryonic stem cell model overexpressing DNMT1. Gene 757, 144934 (2020). •• This report showed for the first time that a number of schizophrenia-associated genes may be dysregulated due to DNMT1 overexpression.
    • 24. Fromer M, Roussos P, Sieberts SK et al. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Nat. Neurosci. 19(11), 1442–1453 (2016).
    • 25. Gandal MJ, Zhang P, Hadjimichael E et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science 362(6420), 1–32 (2019).
    • 26. Jaffe AE, Straub RE, Shin JH et al. Developmental and genetic regulation of the human cortex transcriptome illuminate schizophrenia pathogenesis. Nat. Neurosci. 21(8), 1117–1125 (2019).
    • 27. Saxena S, Choudhury S, Mohan KN. Reproducible differentiation and characterization of neurons from mouse embryonic stem cells. MethodsX 7, 101073 (2020).
    • 28. Saxena S, Choudhury S, Mohan KN. Genome-wide methylation data from R1 (wild-type) and the transgenic DNMT1Tet/Tet mouse embryonic stem cells overexpressing DNA methyltransferase 1 (DNMT1). Data Br. 32, 106242 (2020).
    • 29. Feng H, Conneely KN, Wu H. A Bayesian hierarchical model to detect differentially methylated loci from single nucleotide resolution sequencing data. Nucleic Acids Res. 42(8), 1–11 (2014).
    • 30. Park Y, Wu H. Differential methylation analysis for BS-seq data under general experimental design. Bioinformatics 32(10), 1446–1453 (2016).
    • 31. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method. Methods 25(4), 402–408 (2001).
    • 32. Szklarczyk D, Gable AL, Lyon D et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47(D1), D607–D613 (2019).
    • 33. Xiong Z, Laird PW. COBRA: a sensitive and quantitative DNA methylation assay. Nucleic Acids Res. 25(12), 2532–2534 (1997).
    • 34. D'Haene B, Vandesompele J, Hellemans J. Accurate and objective copy number profiling using real-time quantitative PCR. Methods 50(4), 262–270 (2010).
    • 35. Xu J, Hao Z, Gou X et al. Imaging of reactive oxygen species burst from mitochondria using laser scanning confocal microscopy. Microsc. Res. Tech. 76(6), 612–617 (2013).
    • 36. Borowczyk E, Mohan KN, D'Aiuto L, Cirio MC, Chaillet JR. Identification of a region of the DNMT1 methyltransferase that regulates the maintenance of genomic imprints. Proc. Natl Acad. Sci. USA 106(49), 20806–20811 (2009).
    • 37. Peng Y, Xu Y, Cui D. Wnt signaling pathway in schizophrenia. CNS & Neurological Disorders – Drug Targets 13(5), 755–764 (2014).
    • 38. Funk AJ, McCullumsmith RE, Haroutunian V, Meador-Woodruff JH. Abnormal activity of the MAPK- and cAMP-associated signaling pathways in frontal cortical areas in postmortem brain in schizophrenia. Neuropsychopharmacology 37(4), 896–905 (2012).
    • 39. Berretta S. Extracellular matrix abnormalities in schizophrenia. Neuropharmacology 62(3), 1584–1597 (2012).
    • 40. Wang Z, Li P, Wu T, Zhu S, Deng L, Cui G. Axon guidance pathway genes are associated with schizophrenia risk. Exp. Ther. Med. 16(6), 4519–4526 (2018).
    • 41. Ashton A, Jagannath A. Disrupted sleep and circadian rhythms in schizophrenia and their interaction with dopamine signaling. Front. Neurosci. 14, 1–14 (2020).
    • 42. Kalkman HO. Altered growth factor signaling pathways as the basis of aberrant stem cell maturation in schizophrenia. Pharmacol. Ther. 121(1), 115–122 (2009).
    • 43. Jarskog LF, Glantz LA, Gilmore JH, Lieberman JA. Apoptotic mechanisms in the pathophysiology of schizophrenia. Prog. Neuro-Psychopharmacology Biol. Psychiatry 29(5), 846–858 (2005).
    • 44. Robicsek O, Karry R, Petit I et al. Abnormal neuronal differentiation and mitochondrial dysfunction in hair follicle-derived induced pluripotent stem cells of schizophrenia patients. Mol. Psychiatry 18(10), 1067–1076 (2013).
    • 45. Tucker KL, Beard C, Dausman J et al. Germ-line passage is required for establishment of methylation and expression patterns of imprinted but not of nonimprinted genes. Genes Dev. 10(8), 1008–1020 (1996).
    • 46. Yang YJ, Luo T, Zhao Y et al. Altered insulin-like growth factor-2 signaling is associated with psychopathology and cognitive deficits in patients with schizophrenia. PLoS ONE 15(3), 1–13 (2020).
    • 47. Gardiner E, Beveridge NJ, Wu JQ et al. Imprinted DLK1-DIO3 region of 14q32 defines a schizophrenia-associated miRNA signature in peripheral blood mononuclear cells. Mol. Psychiatry 17(8), 827–840 (2012).
    • 48. Karis K, Eskla KL, Kaare M et al. Altered expression profile of igLON family of neural cell adhesion molecules in the dorsolateral prefrontal cortex of schizophrenic patients. Front. Mol. Neurosci. 11, 1–12 (2018).
    • 49. Crespi B, Read S, Salminen I, Hurd P. A genetic locus for paranoia. Biol. Lett. 14(1), 1–5 (2018).
    • 50. Konopaske GT, Subburaju S, Coyle JT, Benes FM. Altered prefrontal cortical MARCKS and PPP1R9A mRNA expression in schizophrenia and bipolar disorder. Schizophr. Res. 164(1–3), 100–108 (2015).
    • 51. Baylin SB. DNA methylation and gene silencing in cancer. Nat. Clin. Pract. Oncol. 2, 4–11 (2005).
    • 52. Newell-Price J, Clark AJL, King P. DNA methylation and silencing of gene expression. Trends Endocrinol. Metab. 11(4), 142–148 (2000).
    • 53. Clements EG, Mohammad HP, Leadem BR et al. DNMT1 modulates gene expression without its catalytic activity partially through its interactions with histone-modifying enzymes. Nucleic Acids Res. 40(10), 4334–4346 (2012). •• This study provided a mechanism by which DNMT1 can alter gene expression without the involvement of a change in the levels of DNA methylation at the promoters of the target genes.
    • 54. Espada J. Non-catalytic functions of DNMT1. Epigenetics 7(2), 115–118 (2012).
    • 55. Doyle GA, Crist RC, Karatas ET et al. Analysis of LINE-1 elements in DNA from postmortem brains of individuals with schizophrenia. Neuropsychopharmacology 42(13), 2602–2611 (2017).
    • 56. Guffanti G, Gaudi S, Klengel T et al. LINE1 insertions as a genomic risk factor for schizophrenia: preliminary evidence from an affected family. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 171(4), 534–545 (2016).
    • 57. Li S, Yang Q, Hou Y et al. Hypomethylation of LINE-1 elements in schizophrenia and bipolar disorder. J. Psychiatr. Res. 107, 68–72 (2018).
    • 58. Misiak B, Szmida E, Karpiński P, Loska O, Sąsiadek MM, Frydecka D. Lower LINE-1 methylation in first-episode schizophrenia patients with the history of childhood trauma. Epigenomics 7(8), 1275–1285 (2015). •• One of the first reports suggesting that the role of hypomethylation and increased LINE1 burden in conferring risk for schizophrenia.
    • 59. Terry DM, Devine SE. Aberrantly high levels of somatic LINE-1 expression and retro transposition in human neurological disorders. Front. Genet. 10, 1–14 (2020).
    • 60. Kalayasiri R, Kraijak K, Mutirangura A, Maes M. Paranoid schizophrenia and methamphetamine-induced paranoia are both characterized by a similar LINE-1 partial methylation profile, which is more pronounced in paranoid schizophrenia. Schizophr. Res. 208, 221–227 (2019).
    • 61. Shpyleva S, Melnyk S, Pavliv O, Pogribny I, Jill James S. Overexpression of LINE-1 retrotransposons in autism brain. Mol. Neurobiol. 55(2), 1740–1749 (2018).
    • 62. Dong E, Gavin DP, Chen Y, Davis J. Upregulation of TET1 and downregulation of APOBEC3A and APOBEC3C in the parietal cortex of psychotic patients. Transl. Psychiatry 2, 1–7 (2012).
    • 63. Kwon W, Kim HS, Jeong J et al. Tet1 overexpression leads to anxiety-like behavior and enhanced fear memories via the activation of calcium-dependent cascade through Egr1 expression in mice. FASEB J. 32(1), 390–403 (2018).
    • 64. Dong E, Chen Y, Gavin DP, Grayson DR, Guidotti A. Valproate induces DNA demethylation in nuclear extracts from adult mouse brain. Epigenetics 5(8), 730–735 (2010).
    • 65. Smith RC, Lindenmayer JP, Davis JM et al. Cognitive and antismoking effects of varenicline in patients with schizophrenia or schizoaffective disorder. Schizophr. Res. 110(1–3), 149–155 (2009).
    • 66. Matrisciano F, Dong E, Nicoletti F, Guidotti A. Epigenetic alterations in prenatal stress mice as an endophenotype model for schizophrenia: role of metabotropic glutamate 2/3 receptors. Front. Mol. Neurosci. 11, 1–8 (2018).
    • 67. Dong E, Ruzicka WB, Grayson DR, Guidotti A. DNA-methyltransferase1 (DNMT1) binding to CpG rich GABAergic and BDNF promoters is increased in the brain of schizophrenia and bipolar disorder patients. Schizophr. Res. 167(1–3), 35–41 (2015). • One of the early studies that reported the catalytic-independent activity of DNMT1 in regulating the expression of BDNF and RELN genes in absence of a DNA methylation change in the postmortem brain tissues of schizophrenia patients.
    • 68. Miller JL, Grant PA. The role of DNA methylation and histone modifications in transcriptional regulation in humans. Subcell Biochem. 61, 289–317 (2013).
    • 69. Barbot W, Dupressoir A, Lazar V, Heidmann T. Epigenetic regulation of an IAP retrotransposon in the aging mouse: progressive demethylation and de-silencing of the element by its repetitive induction. Nucleic Acids Res. 30(11), 2365–2373 (2002).
    • 70. Lane N, Dean W, Erhardt S et al. Resistance of IAPs to methylation reprogramming may provide a mechanism for epigenetic inheritance in the mouse. Genesis 35(2), 88–93 (2003).
    • 71. Belancio VP, Hedges DJ, Deininger P. LINE-1 RNA splicing and influences on mammalian gene expression. Nucleic Acids Res. 34(5), 1512–1521 (2006).
    • 72. Murata Y, Ikegame T, Koike S et al. Global DNA hypomethylation and its correlation to the betaine level in peripheral blood of patients with schizophrenia. Prog. Neuro-Psychopharmacology Biol. Psychiatry 99, 109855 (2020).
    • 73. Shimabukuro M, Sasaki T, Imamura A et al. Global hypomethylation of peripheral leukocyte DNA in male patients with schizophrenia: a potential link between epigenetics and schizophrenia. J. Psychiatr. Res. 41(12), 1042–1046 (2007). • One of the first reports showing a reduction in the genome-wide methylation levels in schizophrenia.