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

Evaluation of DNA methylation in BDNF, SLC6A4, NR3C1 and FKBP5 before and after treatment with selective serotonin-reuptake inhibitor in major depressive disorder

    Saeid Mohammadi

    Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran

    ,
    Abbas Beh-Pajooh‡

    Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Mahnaz Ahmadimanesh‡

    Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Mohsen Amini

    Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran

    ,
    Mahmoud Ghazi-Khansari

    Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran

    ,
    Seyed Adel Moallem

    Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran

    Department of Pharmacology and Toxicology, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq

    ,
    Rohollah Hosseini

    Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran

    ,
    Yazdan Hasani Nourian

    Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran

    &
    Mohammad Hossein Ghahremani

    *Author for correspondence: Tel.: +98 216 695 9102;

    E-mail Address: mhghahremani@tums.ac.ir

    Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran

    Toxicology and Poisoning Research Center, Tehran University of Medical Sciences, Tehran, Iran

    Published Online:https://doi.org/10.2217/epi-2022-0246

    Aim: To identify the DNA methylation status of related genes in major depressive disorder following selective serotonin-reuptake inhibitor treatment. Materials & methods: 45 patients with major depressive disorder and 45 healthy volunteers were considered experimental and control groups, respectively. High-resolution melting real-time PCR was implemented to evaluate DNA methylation. Results: After 100 days of selective serotonin-reuptake inhibitor treatment, methylation of promoter CpG sites of BDNF, NR3C1, FKBP5 and SLC6A4 was significantly reduced. Compared with before treatment, patients' Hamilton Depression Rating Scale scores were significantly reduced after selective serotonin-reuptake inhibitor treatment (p ≤ 0.0001). Conclusion: Based on the proven effect of antidepressants on DNA methylation and gene expression, these medications can improve the treatment process and reduce depression scores after treatment.

    Graphical abstract

    Overview of pathway connecting risk factors for depression to gene methylation.

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

    References

    • 1. Hardeveld F, Spijker J, De Graaf R, Nolen W, Beekman A. Prevalence and predictors of recurrence of major depressive disorder in the adult population. Acta Psychiatr. Scand. 122(3), 184–191 (2010).
    • 2. Clark SL, Hattab MW, Chan RF et al. A methylation study of long-term depression risk. Mol. Psychiatry 25(6), 1334–1343 (2020).
    • 3. Frokjaer VG. Pharmacological sex hormone manipulation as a risk model for depression. J. Neurosci. Res. 98(7), 1283–1292 (2020).
    • 4. Gharraee B, Tajrishi KZ, Sheybani F et al. Prevalence of major depressive disorder in the general population of Iran: a systematic review and meta-analysis. Med. J. Islam. Repub. Iran 33, 151 (2019).
    • 5. Lee M-S. Pharmacogenetic treatment of depressive disorders Future Neurol. 2(3), 329–338 2007).
    • 6. Kraus C, Kadriu B, Lanzenberger R, Zarate CA Jr, Kasper S. Prognosis and improved outcomes in major depression: a review. Transl. Psychiatry 9(1), 127 (2019).
    • 7. Bialek K, Czarny P, Strycharz J, Sliwinski T. Major depressive disorders accompanying autoimmune diseases – response to treatment. Prog. Neuropsychopharmacol. Biol. Psychiatry 95 109678 (2019).
    • 8. Mullins CD, Shaya FT, Meng F, Wang J, Harrison D. Persistence, switching, and discontinuation rates among patients receiving sertraline, paroxetine, and citalopram. Pharmacotherapy 25(5), 660–667 (2005). • Epigenetic changes are involved in the progression and treatment of depression.
    • 9. Lozupone M, Panza F. Social determinants of late-life depression epigenetics. Epigenomics 12(7), 559–562 (2020).
    • 10. Isbister GK, Bowe SJ, Dawson A, Whyte IM. Relative toxicity of selective serotonin reuptake inhibitors (SSRIs) in overdose. J. Toxicol. Clin. Toxicol. 42(3), 277–285 (2004).
    • 11. Narasimhan S, Lohoff FW. Pharmacogenetics of antidepressant drugs: current clinical practice and future directions. Pharmacogenomics 13(4), 441–464 (2012).
    • 12. Hyttel J,Pharmacological characterization of selective serotonin reuptake inhibitors (SSRIs). Int. Clin. Psychopharmacol. 9( Suppl. 1),19–26( 1994).
    • 13. Reynolds GP, Fachim HA. Does DNA methylation influence the effects of psychiatric drugs? Epigenomics 8(3), 309–312 (2016).
    • 14. Tsai S-J, Hong C-J, Liou Y-J. Brain-derived neurotrophic factor and antidepressant action: another piece of evidence from pharmacogenetics. Pharmacogenomics 9(9),1353–1358( 2008).
    • 15. García-Marín LM, Rabinowitz JA, Ceja Z, Alcauter S, Medina-Rivera A, Rentería ME. The pharmacogenomics of selective serotonin reuptake inhibitors. Pharmacogenomics 4, 233–244( 2022).
    • 16. Czarny P, Białek K, Ziółkowska S, Strycharz J, Barszczewska G, Sliwinski T. The importance of epigenetics in diagnostics and treatment of major depressive disorder. J. Pers. Med. 11(3), 167 (2021).
    • 17. Zhou J, Li M, Wang X et al. Drug response-related DNA methylation changes in schizophrenia, bipolar disorder, and major depressive disorder. Front. Neurosci. 15, 674273( 2021).
    • 18. Kimmel M, Kaminsky Z, Payne JL. Biomarker or pathophysiology? The role of DNA methylation in postpartum depression. Epigenomics 5(5), 473–475 (2013).
    • 19. Volkow ND, Koob G, Baler R. Biomarkers in substance use disorders. ACS Chem. Neurosci. 6(4), 522–525 (2015).
    • 20. Dereix AE, Ledyard R, Redhunt AM et al. Maternal anxiety and depression in pregnancy and DNA methylation of the NR3C1 glucocorticoid receptor gene. Epigenomics 13(21), 1701–1709 (2021).
    • 21. Shields AE, Wise LA, Ruiz-Narvaez EA et al. Childhood abuse, promoter methylation of leukocyte NR3C1 and the potential modifying effect of emotional support. Epigenomics 8(11), 1507–1517 (2016).
    • 22. Nagarajan S, Seddighzadeh B, Baccarelli A, Wise LA, Williams M, Shields AE. Adverse maternal exposures, methylation of glucocorticoid-related genes and perinatal outcomes: a systematic review. Epigenomics 8(7), 925–944 (2016).
    • 23. Winkler BK, Lehnert H, Oster H, Kirchner H, Harbeck B. FKBP5 methylation as a possible marker for cortisol state and transient cortisol exposure in healthy human subjects. Epigenomics 9(10), 1279–1286 (2017).
    • 24. Montirosso R, Provenzi L, Giorda R et al. SLC6A4 promoter region methylation and socio-emotional stress response in very preterm and full-term infants. Epigenomics 8(7), 895–907 (2016).
    • 25. Park C, Rosenblat JD, Brietzke E et al. Stress, epigenetics and depression: a systematic review. Neurosci. Biobehav. Rev. 102, 139–152 (2019).
    • 26. Iamjan S-A, Thanoi S, Watiktinkorn P et al. Changes of BDNF exon IV DNA methylation are associated with methamphetamine dependence. Epigenomics 13(12), 953–965 (2021).
    • 27. Scharinger C, Bartova L, Pezawas L. Brain-derived neurotrophic factor: a peripheral biomarker for major depressive disorder and antidepressant efficacy? Per. Med. 8(2), 115–117 (2011).
    • 28. Ahmadimanesh M, Abbaszadegan MR, Rad DM et al. Effects of selective serotonin reuptake inhibitors on DNA damage in patients with depression. J. Psychopharmacol. 33(11), 1364–1376 (2019).
    • 29. Switzeny OJ, Christmann M, Renovanz M, Giese A, Sommer C, Kaina B. MGMT promoter methylation determined by HRM in comparison to MSP and pyrosequencing for predicting high-grade glioma response. Clin. Epigenetics 8(1), 1–10 (2016).
    • 30. Nourian YH, Beh-Pajooh A, Aliomrani M et al. Changes in DNA methylation in APOE and ACKR3 genes in multiple sclerosis patients and the relationship with their heavy metal blood levels. Neurotoxicology 87, 182–187 (2021).
    • 31. Numakawa T, Suzuki S, Kumamaru E, Adachi N, Richards M, Kunugi H. BDNF function and intracellular signaling in neurons. Histol. Histopathol. 25, 237–258(2010).
    • 32. Stein DJ, Daniels WM, Savitz J, Harvey BH. Brain-derived neurotrophic factor: the neurotrophin hypothesis of psychopathology. CNS Spectr. 13(11), 945–949 (2008).
    • 33. Castrén E, Rantamäki T. The role of BDNF and its receptors in depression and antidepressant drug action: reactivation of developmental plasticity. Dev. Neurobiol. 70(5), 289–297 (2010).
    • 34. Aid T, Kazantseva A, Piirsoo M, Palm K, Timmusk T. Mouse and rat BDNF gene structure and expression revisited. J. Neurosci. Res. 85(3), 525–535 (2007).
    • 35. Martinowich K, Hattori D, Wu H et al. DNA methylation-related chromatin remodeling in activity-dependent BDNF gene regulation. Science 302(5646), 890–893 (2003). •• Among depressed individuals, DNA methylation is higher in the BDNF promoter.
    • 36. Fuchikami M, Morinobu S, Segawa M et al. DNA methylation profiles of the brain-derived neurotrophic factor (BDNF) gene as a potent diagnostic biomarker in major depression. PLOS ONE 6(8), e23881 (2011).
    • 37. Tadić A, Müller-Engling L, Schlicht K et al. Methylation of the promoter of brain-derived neurotrophic factor exon IV and antidepressant response in major depression. Mol. Psychiatry 19(3), 281–283 (2014).
    • 38. Chen D, Meng L, Pei F, Zheng Y, Leng J. A review of DNA methylation in depression. J. Clin. Neurosci. 43, 39–46 (2017). •• DNA methylation changes in BDNF gene in response to paroxetine treatment.
    • 39. Gassen NC, Fries GR, Zannas AS et al. Chaperoning epigenetics: FKBP51 decreases the activity of DNMT1 and mediates epigenetic effects of the antidepressant paroxetine. Sci. Signal. 8(404), ra119 (2015).
    • 40. Melas PA, Rogdaki M, Lennartsson A et al. Antidepressant treatment is associated with epigenetic alterations in the promoter of P11 in a genetic model of depression. Int. J. Neuropsychopharmacol. 15(5), 669–679 (2012).
    • 41. Zhang Y, Gu F, Chen J, Dong W. Chronic antidepressant administration alleviates frontal and hippocampal BDNF deficits in CUMS rat. Brain Res. 1366, 141–148 (2010).
    • 42. Russo-Neustadt A, Ha T, Ramirez R, Kesslak JP. Physical activity–antidepressant treatment combination: impact on brain-derived neurotrophic factor and behavior in an animal model. Behav. Brain Res. 120(1), 87–95 (2001).
    • 43. Turner JD, Alt SR, Cao L et al. Transcriptional control of the glucocorticoid receptor: CpG islands, epigenetics and more. Biochem. Pharmacol. 80(12), 1860–1868 (2010). •• NR3C1 methylation of DNA increases in infants of depressed mothers.
    • 44. Murgatroyd C, Quinn J, Sharp H, Pickles A, Hill J. Effects of prenatal and postnatal depression, and maternal stroking, at the glucocorticoid receptor gene. Transl. Psychiatry 5(5), e560 (2015).
    • 45. Oberlander TF, Weinberg J, Papsdorf M, Grunau R, Misri S, Devlin AM. Prenatal exposure to maternal depression, neonatal methylation of human glucocorticoid receptor gene (NR3C1) and infant cortisol stress responses. Epigenetics 3(2), 97–106 (2008).
    • 46. Luddington NS, Mandadapu A, Husk M, El-Mallakh RS. Clinical implications of genetic variation in the serotonin transporter promoter region: a review. Prim. Care Companion J. Clin. Psychiatry 11(3), 93 (2009).
    • 47. Webb LM, Phillips KE, Ho MC, Veldic M, Blacker CJ. The relationship between DNA methylation and antidepressant medications: a systematic review. Int. J. Mol. Sci. 21(3), 826 (2020).
    • 48. Philibert R, Madan A, Andersen A, Cadoret R, Packer H, Sandhu H. Serotonin transporter mRNA levels are associated with the methylation of an upstream CpG island. Am. J. Med. Genet. B Neuropsychiatr. Genet. 144(1), 101–105 (2007).
    • 49. Olsson C, Foley D, Parkinson-Bates M et al. Prospects for epigenetic research within cohort studies of psychological disorder: a pilot investigation of a peripheral cell marker of epigenetic risk for depression. Biol. Psychol. 83(2), 159–165 (2010).
    • 50. Gaspar P, Cases O, Maroteaux L. The developmental role of serotonin: news from mouse molecular genetics. Nat. Rev. Neurosci. 4(12), 1002–1012 (2003).
    • 51. Schiele MA, Zwanzger P, Schwarte K, Arolt V, Baune BT, Domschke K. Serotonin transporter gene promoter hypomethylation as a predictor of antidepressant treatment response in major depression: a replication study. Int. J. Neuropsychopharmacol. 24(3), 191–199 (2021).
    • 52. 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. 225, 422–428 (2018).
    • 53. Zannas AS, Wiechmann T, Gassen NC, Binder EB. Gene–stress–epigenetic regulation of FKBP5: clinical and translational implications. Neuropsychopharmacology 41(1), 261–274 (2016). •• DNA methylation in the glucocorticoid receptor (GR) system is associated with depression.
    • 54. Höhne N, Poidinger M, Merz F et al. FKBP5 genotype-dependent DNA methylation and mRNA regulation after psychosocial stress in remitted depression and healthy controls. Int. J. Neuropsychopharmacol. 18(4), pyu087 (2015).
    • 55. Weder N, Zhang H, Jensen K et al. Child abuse, depression, and methylation in genes involved with stress, neural plasticity, and brain circuitry. J. Am. Acad. Child Adolesc. Psychiatry 53(4), 417–424.e5 (2014).
    • 56. 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. Neuropsychopharmacology 44(5), 930–938 (2019).
    • 57. Miller O, Shakespeare-Finch J, Bruenig D, Mehta D. DNA methylation of NR3C1 and FKBP5 is associated with posttraumatic stress disorder, posttraumatic growth, and resilience. Psychol. Trauma 12(7), 750 (2020).
    • 58. Matosin N, Halldorsdottir T, Binder EB. Understanding the molecular mechanisms underpinning gene by environment interactions in psychiatric disorders: the FKBP5 model. Biol. Psychiatry 83(10), 821–830 (2018).
    • 59. Yehuda R, Daskalakis NP, Desarnaud F et al. Epigenetic biomarkers as predictors and correlates of symptom improvement following psychotherapy in combat veterans with PTSD. Front. Psychiatry 4(118), (2013).
    • 60. Lopez J, Mamdani F, Labonte B et al. Epigenetic regulation of BDNF expression according to antidepressant response. Mol. Psychiatry 18(4), 398–399 (2013).
    • 61. Okada S, Morinobu S, Fuchikami M et al. The potential of SLC6A4 gene methylation analysis for the diagnosis and treatment of major depression. J. Psychiatr. Res. 53, 47–53 (2014).