Hostname: page-component-848d4c4894-p2v8j Total loading time: 0.001 Render date: 2024-05-26T21:21:05.031Z Has data issue: false hasContentIssue false

Neuroimaging and epigenetic analysis reveal novel epigenetic loci in major depressive disorder

Published online by Cambridge University Press:  09 May 2024

Hyun-Ho Yang
Affiliation:
Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
Kyu-Man Han
Affiliation:
Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea Brain Convergence Research Center, Korea University College of Medicine, Seoul, Republic of Korea
Aram Kim
Affiliation:
Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
Youbin Kang
Affiliation:
Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
Woo-Suk Tae
Affiliation:
Brain Convergence Research Center, Korea University College of Medicine, Seoul, Republic of Korea
Mi-Ryung Han*
Affiliation:
Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
Byung-Joo Ham*
Affiliation:
Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea Brain Convergence Research Center, Korea University College of Medicine, Seoul, Republic of Korea
*
Corresponding author: Mi-Ryung Han; Email: genetic0309@inu.ac.kr; Byung-Joo Ham; Email: hambj@korea.ac.kr
Corresponding author: Mi-Ryung Han; Email: genetic0309@inu.ac.kr; Byung-Joo Ham; Email: hambj@korea.ac.kr

Abstract

Background

Epigenetic modifications, such as DNA methylation, contribute to the pathophysiology of major depressive disorder (MDD). This study aimed to identify novel MDD-associated epigenetic loci using DNA methylation profiles and explore the correlations between epigenetic loci and cortical thickness changes in patients with MDD.

Methods

A total of 350 patients with MDD and 161 healthy controls (HCs) were included in the epigenome-wide association studies (EWAS). We analyzed methylation, copy number alteration (CNA), and gene network profiles in the MDD group. A total of 234 patients with MDD and 135 HCs were included in neuroimaging methylation analysis. Pearson's partial correlation analysis was used to estimate the correlation between cortical thickness of brain regions and DNA methylation levels of the loci.

Results

In total, 2018 differentially methylated probes (DMPs) and 351 differentially methylated regions (DMRs) were identified. DMP-related genes were enriched in two networks involved in the central nervous system. In neuroimaging analysis, patients with MDD showed cortical thinning in the prefrontal regions and cortical thickening in several occipital regions. Cortical thickness of the left ventrolateral prefrontal cortex (VLPFC, i.e. pars triangularis) was negatively correlated with eight DMPs associated with six genes (EML6, ZFP64, CLSTN3, KCNMA1, TAOK2, and NT5E).

Conclusion

Through combining DNA methylation and neuroimaging analyses, negative correlations were identified between the cortical thickness of the left VLPFC and DNA methylation levels of eight DMPs. Our findings could improve our understanding of the pathophysiology of MDD.

Type
Original Article
Copyright
Copyright © The Author(s), 2024. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Footnotes

*

The first two authors have contributed equally to this article as co-first authors.

References

Aberg, K. A., Dean, B., Shabalin, A. A., Chan, R. F., Han, L. K. M., Zhao, M., … van den Oord, E. (2020). Methylome-wide association findings for major depressive disorder overlap in blood and brain and replicate in independent brain samples. Molecular Psychiatry, 25(6), 13441354. doi:10.1038/s41380-018-0247-6CrossRefGoogle ScholarPubMed
Ables, J. L., Breunig, J. J., Eisch, A. J., & Rakic, P. (2011). Not(ch) just development: Notch signalling in the adult brain. Nature Reviews Neuroscience, 12(5), 269283. doi:10.1038/nrn3024CrossRefGoogle ScholarPubMed
Ambalavanan, A., Girard, S. L., Ahn, K., Zhou, S., Dionne-Laporte, A., Spiegelman, D., … Rouleau, G. A. (2016). De novo variants in sporadic cases of childhood onset schizophrenia. European Journal of Human Genetics, 24(6), 944948. doi:10.1038/ejhg.2015.218CrossRefGoogle ScholarPubMed
Bardai, F. H., & D'Mello, S. R. (2011). Selective toxicity by HDAC3 in neurons: Regulation by Akt and GSK3beta. Journal of Neuroscience, 31(5), 17461751. doi:10.1523/JNEUROSCI.5704-10.2011CrossRefGoogle ScholarPubMed
Barnes, J., Mondelli, V., & Pariante, C. M. (2017). Genetic contributions of inflammation to depression. Neuropsychopharmacology, 42(1), 8198. doi:10.1038/npp.2016.169CrossRefGoogle Scholar
Bédard, A., Tremblay, P., Chernomoretz, A., & Vallières, L. (2007). Identification of genes preferentially expressed by microglia and upregulated during cuprizone-induced inflammation. Glia, 55(8), 777789. doi:10.1002/glia.20477CrossRefGoogle ScholarPubMed
Bigdeli, T. B., Maher, B. S., Zhao, Z., Sun, J., Medeiros, H., Akula, N., … Fanous, A. H. (2013). Association study of 83 candidate genes for bipolar disorder in chromosome 6q selected using an evidence-based prioritization algorithm. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics, 162b(8), 898906. doi:10.1002/ajmg.b.32200CrossRefGoogle ScholarPubMed
Cattarinussi, G., Delvecchio, G., Sambataro, F., & Brambilla, P. (2022). The effect of polygenic risk scores for major depressive disorder, bipolar disorder and schizophrenia on morphological brain measures: A systematic review of the evidence. Journal of Affective Disorders, 310, 213222. doi:10.1016/j.jad.2022.05.007CrossRefGoogle Scholar
Chapman, N. H., Nato, A. Q., Bernier, R., Ankenman, K., Sohi, H., Munson, J., … Wijsman, E. M. (2015). Whole exome sequencing in extended families with autism spectrum disorder implicates four candidate genes. Human Genetics, 134(10), 10551068. doi:10.1007/s00439-015-1585-yCrossRefGoogle ScholarPubMed
Cheng, P., Qiu, Z., & Du, Y. (2021). Potassium channels and autism spectrum disorder: An overview. International Journal of Developmental Neuroscience, 81(6), 479491. doi:10.1002/jdn.10123CrossRefGoogle ScholarPubMed
Chiarella, J., Schumann, L., Pomares, F. B., Frodl, T., Tozzi, L., Nemoda, Z., … Booij, L. (2020). DNA methylation differences in stress-related genes, functional connectivity and gray matter volume in depressed and healthy adolescents. Journal of Affective Disorders, 271, 160168. doi:10.1016/j.jad.2020.03.062CrossRefGoogle ScholarPubMed
Choi, H.-K., Choi, Y., Kang, H., Lim, E.-J., Park, S.-Y., Lee, H.-S., … Yoon, H.-G. (2015). PINK1 positively regulates HDAC3 to suppress dopaminergic neuronal cell death. Human Molecular Genetics, 24(4), 11271141. doi:10.1093/hmg/ddu526CrossRefGoogle ScholarPubMed
Cukkemane, A., Becker, N., Zielinski, M., Frieg, B., Lakomek, N. A., Heise, H., … Weiergräber, O. H. (2021). Conformational heterogeneity coupled with β-fibril formation of a scaffold protein involved in chronic mental illnesses. Translational Psychiatry, 11(1), 639. doi:10.1038/s41398-021-01765-1CrossRefGoogle ScholarPubMed
Cunha, R. A. (2016). How does adenosine control neuronal dysfunction and neurodegeneration? Journal of Neurochemistry, 139(6), 10191055. doi:10.1111/jnc.13724CrossRefGoogle ScholarPubMed
Dale, A. M., Fischl, B., & Sereno, M. I. (1999). Cortical surface-based analysis. I. Segmentation and surface reconstruction. Neuroimage, 9(2), 179194. doi:10.1006/nimg.1998.0395CrossRefGoogle ScholarPubMed
Datta, D., Arion, D., Corradi, J. P., & Lewis, D. A. (2015). Altered expression of CDC42 signaling pathway components in cortical layer 3 pyramidal cells in schizophrenia. Biological Psychiatry, 78(11), 775785. doi:10.1016/j.biopsych.2015.03.030CrossRefGoogle ScholarPubMed
Davies, M. N., Volta, M., Pidsley, R., Lunnon, K., Dixit, A., Lovestone, S., … Mill, J. (2012). Functional annotation of the human brain methylome identifies tissue-specific epigenetic variation across brain and blood. Genome Biology, 13(6), R43. doi:10.1186/gb-2012-13-6-r43CrossRefGoogle ScholarPubMed
de Baumont, A., Maschietto, M., Lima, L., Carraro, D. M., Olivieri, E. H., Fiorini, A., … Brentani, H. (2015). Innate immune response is differentially dysregulated between bipolar disease and schizophrenia. Schizophrenia Research, 161(2–3), 215221. doi:10.1016/j.schres.2014.10.055CrossRefGoogle ScholarPubMed
Destrieux, C., Fischl, B., Dale, A., & Halgren, E. (2010). Automatic parcellation of human cortical gyri and sulci using standard anatomical nomenclature. Neuroimage, 53(1), 115. doi:10.1016/j.neuroimage.2010.06.010CrossRefGoogle ScholarPubMed
Dresselhaus, E. C., & Meffert, M. K. (2019). Cellular specificity of NF-κB function in the nervous system. Frontiers in Immunology, 10, 1043. doi:10.3389/fimmu.2019.01043CrossRefGoogle ScholarPubMed
Duman, R. S., Aghajanian, G. K., Sanacora, G., & Krystal, J. H. (2016). Synaptic plasticity and depression: New insights from stress and rapid-acting antidepressants. Nature Medicine, 22(3), 238249. doi:10.1038/nm.4050CrossRefGoogle ScholarPubMed
Efstathopoulos, P., Andersson, F., Melas, P. A., Yang, L. L., Villaescusa, J. C., Rȕegg, J., … Lavebratt, C. (2018). NR3C1 Hypermethylation in depressed and bullied adolescents. Translational Psychiatry, 8(1), 121. doi:10.1038/s41398-018-0169-8CrossRefGoogle ScholarPubMed
Ferrer, A., Labad, J., Salvat-Pujol, N., Barrachina, M., Costas, J., Urretavizcaya, M., … Soria, V. (2019). BDNF genetic variants and methylation: Effects on cognition in major depressive disorder. Translational Psychiatry, 9(1), 265. doi:10.1038/s41398-019-0601-8CrossRefGoogle ScholarPubMed
First, M. B., Williams, J. B. W., Karg, R. S., & Spitzer, R. L. (2016). SCID-5-CV: Structured clinical interview for DSM-5 disorders: Clinician version. Arlington, VA: American Psychiatric Association Publishing.Google Scholar
Fischl, B., Liu, A., & Dale, A. M. (2001). Automated manifold surgery: Constructing geometrically accurate and topologically correct models of the human cerebral cortex. IEEE Transactions on Medical Imaging, 20(1), 7080. doi:10.1109/42.906426CrossRefGoogle ScholarPubMed
Fischl, B., Salat, D. H., Busa, E., Albert, M., Dieterich, M., Haselgrove, C., … Dale, A. M. (2002). Whole brain segmentation: Automated labeling of neuroanatomical structures in the human brain. Neuron, 33(3), 341355. doi:10.1016/s0896-6273(02)00569-xCrossRefGoogle ScholarPubMed
Fischl, B., Sereno, M. I., & Dale, A. M. (1999). Cortical surface-based analysis. II: Inflation, flattening, and a surface-based coordinate system. Neuroimage, 9(2), 195207. doi:10.1006/nimg.1998.0396CrossRefGoogle Scholar
Fischl, B., van der Kouwe, A., Destrieux, C., Halgren, E., Ségonne, F., Salat, D. H., … Dale, A. M. (2004). Automatically parcellating the human cerebral cortex. Cerebral Cortex, 14(1), 1122. doi:10.1093/cercor/bhg087CrossRefGoogle ScholarPubMed
Flint, J., & Kendler, K. S. (2014). The genetics of major depression. Neuron, 81(3), 484503. doi:10.1016/j.neuron.2014.01.027CrossRefGoogle ScholarPubMed
Freytag, V., Carrillo-Roa, T., Milnik, A., Sämann, P. G., Vukojevic, V., Coynel, D., … Papassotiropoulos, A. (2017). A peripheral epigenetic signature of immune system genes is linked to neocortical thickness and memory. Nature Communications, 8, 15193. doi:10.1038/ncomms15193CrossRefGoogle Scholar
Fujisawa, T. X., Nishitani, S., Takiguchi, S., Shimada, K., Smith, A. K., & Tomoda, A. (2019). Oxytocin receptor DNA methylation and alterations of brain volumes in maltreated children. Neuropsychopharmacology, 44(12), 20452053. doi:10.1038/s41386-019-0414-8CrossRefGoogle ScholarPubMed
Gao, T.-T., Wang, Y., Liu, L., Wang, J.-L., Wang, Y.-J., Guan, W., … Jiang, B. (2020). LIMK1/2 in the mPFC plays a role in chronic stress-induced depressive-like effects in mice. International Journal of Neuropsychopharmacology, 23(12), 821836. doi:10.1093/ijnp/pyaa067CrossRefGoogle Scholar
Gibitova, E. A., Dobrynin, P. V., Pomerantseva, E. A., Musatova, E. V., Kostareva, A., Evsyukov, I., … Grigorenko, E. L. (2022). A study of the genomic variations associated with autistic spectrum disorders in a Russian cohort of patients using whole-exome sequencing. Genes, 13(5), 920. doi:10.3390/genes13050920CrossRefGoogle Scholar
Glahn, D. C., Curran, J. E., Winkler, A. M., Carless, M. A., Kent, J. W. Jr., Charlesworth, J. C., … Blangero, J. (2012). High dimensional endophenotype ranking in the search for major depression risk genes. Biological Psychiatry, 71(1), 614. doi:10.1016/j.biopsych.2011.08.022CrossRefGoogle ScholarPubMed
Gonzales, E. L., Jeon, S. J., Han, K. M., Yang, S. J., Kim, Y., Remonde, C. G., … Shin, C. Y. (2023). Correlation between immune-related genes and depression-like features in an animal model and in humans. Brain, Behavior, and Immunity, 113, 2943. doi:10.1016/j.bbi.2023.06.017CrossRefGoogle Scholar
Gough, S. C. L., & Simmonds, M. J. (2007). The HLA region and autoimmune disease: Associations and mechanisms of action. Current Genomics, 8(7), 453465. doi:10.2174/138920207783591690Google ScholarPubMed
Graw, S., Henn, R., Thompson, J. A., & Koestler, D. C. (2019). pwrEWAS: A user-friendly tool for comprehensive power estimation for epigenome wide association studies (EWAS). BMC Bioinformatics, 20(1), 218. doi:10.1186/s12859-019-2804-7CrossRefGoogle ScholarPubMed
Gunnersen, J. M., Kim, M. H., Fuller, S. J., De Silva, M., Britto, J. M., Hammond, V. E., … Tan, S.-S. (2007). Sez-6 proteins affect dendritic arborization patterns and excitability of cortical pyramidal neurons. Neuron, 56(4), 621639. doi:10.1016/j.neuron.2007.09.018CrossRefGoogle ScholarPubMed
Hall, L. S., Adams, M. J., Arnau-Soler, A., Clarke, T.-K., Howard, D. M., Zeng, Y., … McIntosh, A. M. (2018). Genome-wide meta-analyses of stratified depression in Generation Scotland and UK Biobank. Translational Psychiatry, 8(1), 112. doi:10.1038/s41398-017-0034-1CrossRefGoogle ScholarPubMed
Hamilton, M. (1960). A rating scale for depression. Journal of Neurology, Neurosurgery, and Psychiatry, 23(1), 5662.CrossRefGoogle ScholarPubMed
Han, K. M., Choi, K. W., Kim, A., Kang, W., Kang, Y., Tae, W. S., … Ham, B. J. (2022). Association of DNA methylation of the NLRP3 gene with changes in cortical thickness in major depressive disorder. International Journal of Molecular Sciences, 23(10), 5768. doi:10.3390/ijms23105768CrossRefGoogle ScholarPubMed
Han, K. M., & Ham, B. J. (2021). How inflammation affects the brain in depression: A review of functional and structural MRI studies. Journal of Clinical Neurology, 17(4), 503515. doi:10.3988/jcn.2021.17.4.503CrossRefGoogle ScholarPubMed
Han, K. M., Han, M. R., Kim, A., Kang, W., Kang, Y., Kang, J., … Ham, B. J. (2020a). A study combining whole-exome sequencing and structural neuroimaging analysis for major depressive disorder. Journal of Affective Disorders, 262, 3139. doi:10.1016/j.jad.2019.10.039CrossRefGoogle ScholarPubMed
Han, K. M., Tae, W. S., Kim, A., Kang, Y., Kang, W., Kang, J., … Ham, B. J. (2020b). Serum FAM19A5 levels: A novel biomarker for neuroinflammation and neurodegeneration in major depressive disorder. Brain, Behavior, and Immunity, 87, 852859. doi:10.1016/j.bbi.2020.03.021CrossRefGoogle Scholar
Hibar, D. P., Stein, J. L., Renteria, M. E., Arias-Vasquez, A., Desrivières, S., Jahanshad, N., … Medland, S. E. (2015). Common genetic variants influence human subcortical brain structures. Nature, 520(7546), 224229. doi:10.1038/nature14101CrossRefGoogle ScholarPubMed
Hill, W. D., Marioni, R. E., Maghzian, O., Ritchie, S. J., Hagenaars, S. P., McIntosh, A. M., … Deary, I. J. (2019). A combined analysis of genetically correlated traits identifies 187 loci and a role for neurogenesis and myelination in intelligence. Molecular Psychiatry, 24(2), 169181. doi:10.1038/s41380-017-0001-5CrossRefGoogle Scholar
Hogstrom, L. J., Westlye, L. T., Walhovd, K. B., & Fjell, A. M. (2013). The structure of the cerebral cortex across adult life: Age-related patterns of surface area, thickness, and gyrification. Cerebral Cortex, 23(11), 25212530. doi:10.1093/cercor/bhs231CrossRefGoogle ScholarPubMed
Horvath, S., Zhang, Y., Langfelder, P., Kahn, R. S., Boks, M. P. M., van Eijk, K., … Ophoff, R. A. (2012). Aging effects on DNA methylation modules in human brain and blood tissue. Genome Biology, 13(10), R97. doi:10.1186/gb-2012-13-10-r97CrossRefGoogle ScholarPubMed
Hoseth, E. Z., Krull, F., Dieset, I., Mørch, R. H., Hope, S., Gardsjord, E. S., … Ueland, T. (2018). Attenuated Notch signaling in schizophrenia and bipolar disorder. Scientific Reports, 8(1), 5349. doi:10.1038/s41598-018-23703-wCrossRefGoogle ScholarPubMed
Howes, O. D., & Onwordi, E. C. (2023). The synaptic hypothesis of schizophrenia version III: A master mechanism. Molecular Psychiatry, 28(5), 18431856. doi:10.1038/s41380-023-02043-wCrossRefGoogle Scholar
Hu, H. T., Huang, T. N., & Hsueh, Y. P. (2020). KLHL17/Actinfilin, a brain-specific gene associated with infantile spasms and autism, regulates dendritic spine enlargement. Journal of Biomedical Science, 27(1), 103. doi:10.1186/s12929-020-00696-1CrossRefGoogle ScholarPubMed
Huang, Y. S., Fang, T. H., Kung, B., & Chen, C. H. (2022). Two genetic mechanisms in Two siblings with intellectual disability, autism spectrum disorder, and psychosis. Journal of Personalized Medicine, 12(6), 1013. doi:10.3390/jpm12061013CrossRefGoogle ScholarPubMed
Humphreys, K. L., Moore, S. R., Davis, E. G., MacIsaac, J. L., Lin, D. T. S., Kobor, M. S., & Gotlib, I. H. (2019). DNA methylation of HPA-axis genes and the onset of major depressive disorder in adolescent girls: A prospective analysis. Translational Psychiatry, 9(1), 245. doi:10.1038/s41398-019-0582-7CrossRefGoogle ScholarPubMed
Ignatov, A., Lintzel, J., Hermans-Borgmeyer, I., Kreienkamp, H.-J., Joost, P., Thomsen, S., … Schaller, H. C. (2003). Role of the G-protein-coupled receptor GPR12 as high-affinity receptor for sphingosylphosphorylcholine and its expression and function in brain development. Journal of Neuroscience, 23(3), 907914. doi:10.1523/JNEUROSCI.23-03-00907.2003CrossRefGoogle ScholarPubMed
Januar, V., Ancelin, M. L., Ritchie, K., Saffery, R., & Ryan, J. (2015). BDNF promoter methylation and genetic variation in late-life depression. Translational Psychiatry, 5(8), e619. doi:10.1038/tp.2015.114CrossRefGoogle ScholarPubMed
Johnson, W. E., Li, C., & Rabinovic, A. (2007). Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics (Oxford, England), 8(1), 118127. doi:10.1093/biostatistics/kxj037CrossRefGoogle ScholarPubMed
Kajiwara, Y., McKenzie, A., Dorr, N., Gama Sosa, M. A., Elder, G., Schmeidler, J., … Buxbaum, J. D. (2016). The human-specific CASP4 gene product contributes to Alzheimer-related synaptic and behavioural deficits. Human Molecular Genetics, 25(19), 43154327. doi:10.1093/hmg/ddw265CrossRefGoogle ScholarPubMed
Kajiwara, Y., Schiff, T., Voloudakis, G., Gama Sosa, M. A., Elder, G., Bozdagi, O., & Buxbaum, J. D. (2014). A critical role for human caspase-4 in endotoxin sensitivity. The Journal of Immunology, 193(1), 335343. doi:10.4049/jimmunol.1303424CrossRefGoogle ScholarPubMed
Kang, H. J., Kim, J. M., Stewart, R., Kim, S. Y., Bae, K. Y., Kim, S. W., … Yoon, J. S. (2013). Association of SLC6A4 methylation with early adversity, characteristics and outcomes in depression. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 44, 2328. doi:10.1016/j.pnpbp.2013.01.006CrossRefGoogle ScholarPubMed
Kang, H. J., Kim, K. T., Yoo, K. H., Park, Y., Kim, J. W., Kim, S. W., … Kim, J. M. (2020). Genetic markers for later remission in response to early improvement of antidepressants. International Journal of Molecular Sciences, 21(14), 4884. doi:10.3390/ijms21144884CrossRefGoogle ScholarPubMed
Kang, M., Ang, T. F. A., Devine, S. A., Sherva, R., Mukherjee, S., Trittschuh, E. H., … Farrer, L. A. (2023). A genome-wide search for pleiotropy in more than 100000 harmonized longitudinal cognitive domain scores. Molecular Neurodegeneration, 18(1), 40. doi:10.1186/s13024-023-00633-4CrossRefGoogle Scholar
Kaufman, J., Wymbs, N. F., Montalvo-Ortiz, J. L., Orr, C., Albaugh, M. D., Althoff, R., … Hudziak, J. (2018). Methylation in OTX2 and related genes, maltreatment, and depression in children. Neuropsychopharmacology, 43(11), 22042211. doi:10.1038/s41386-018-0157-yCrossRefGoogle ScholarPubMed
Kaur, A., Lee, L.-H., Chow, S.-C., & Fang, C.-M. (2018). IRF5-mediated Immune responses and its implications in immunological disorders. International Reviews of Immunology, 37(5), 229248. doi:10.1080/08830185.2018.1469629CrossRefGoogle ScholarPubMed
Kessler, R. C., Angermeyer, M., Anthony, J. C., Deg, R., Demyttenaere, K., Gasquet, I., … Ustün, T. B. (2007). Lifetime prevalence and age-of-onset distributions of mental disorders in the world health organization's world mental health survey initiative. World Psychiatry, 6(3), 168176.Google ScholarPubMed
Kim, Y. K., Ham, B. J., & Han, K. M. (2019). Interactive effects of genetic polymorphisms and childhood adversity on brain morphologic changes in depression. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 91, 413. doi:10.1016/j.pnpbp.2018.03.009CrossRefGoogle ScholarPubMed
Klengel, T., & Binder, E. B. (2013). Gene–environment interactions in major depressive disorder. The Canadian Journal of Psychiatry, 58(2), 7683. doi:10.1177/070674371305800203CrossRefGoogle ScholarPubMed
Klinger-König, J., Hertel, J., Van der Auwera, S., Frenzel, S., Pfeiffer, L., Waldenberger, M., … Grabe, H. J. (2019). Methylation of the FKBP5 gene in association with FKBP5 genotypes, childhood maltreatment and depression. Neuropsychopharmacology, 44(5), 930938. doi:10.1038/s41386-019-0319-6CrossRefGoogle ScholarPubMed
Kupfer, D. J., Frank, E., & Phillips, M. L. (2012). Major depressive disorder: New clinical, neurobiological, and treatment perspectives. Lancet (London, England), 379(9820), 10451055. doi:10.1016/s0140-6736(11)60602-8CrossRefGoogle ScholarPubMed
Lapato, D. M., Roberson-Nay, R., Kirkpatrick, R. M., Webb, B. T., York, T. P., & Kinser, P. A. (2019). DNA methylation associated with postpartum depressive symptoms overlaps findings from a genome-wide association meta-analysis of depression. Clinical Epigenetics, 11(1), 169. doi:10.1186/s13148-019-0769-zCrossRefGoogle ScholarPubMed
Laumonnier, F., Roger, S., Guérin, P., Molinari, F., M'Rad, R., Cahard, D., … Briault, S. (2006). Association of a functional deficit of the BKCa channel, a synaptic regulator of neuronal excitability, with autism and mental retardation. American Journal of Psychiatry, 163(9), 16221629. doi:10.1176/ajp.2006.163.9.1622CrossRefGoogle Scholar
Laurent, L., Wong, E., Li, G., Huynh, T., Tsirigos, A., Ong, C. T., … Wei, C.-L. (2010). Dynamic changes in the human methylome during differentiation. Genome Research, 20(3), 320331. doi:10.1101/gr.101907.109CrossRefGoogle ScholarPubMed
Lee, J. J., Wedow, R., Okbay, A., Kong, E., Maghzian, O., Zacher, M., … Cesarini, D. (2018). Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nature Genetics, 50(8), 11121121. doi:10.1038/s41588-018-0147-3CrossRefGoogle ScholarPubMed
Lennon, P. F., Taylor, C. T., Stahl, G. L., & Colgan, S. P. (1998). Neutrophil-derived 5′-adenosine monophosphate promotes endothelial barrier function via CD73-mediated conversion to adenosine and endothelial A2B receptor activation. The Journal of Experimental Medicine, 188(8), 14331443.CrossRefGoogle ScholarPubMed
Li, M., Li, Y., Qin, H., Tubbs, J. D., Li, M., Qiao, C., … Yao, Y. (2021). Genome-wide DNA methylation analysis of peripheral blood cells derived from patients with first-episode schizophrenia in the Chinese Han population. Molecular Psychiatry, 26(8), 44754485. doi:10.1038/s41380-020-00968-0CrossRefGoogle ScholarPubMed
Li, Q. S., Morrison, R. L., Turecki, G., & Drevets, W. C. (2022). Meta-analysis of epigenome-wide association studies of major depressive disorder. Scientific Reports, 12(1), 18361. doi:10.1038/s41598-022-22744-6CrossRefGoogle ScholarPubMed
Liu, Y.-J., Chen, J., Li, X., Zhou, X., Hu, Y.-M., Chu, S.-F., … Chen, N.-H. (2019). Research progress on adenosine in central nervous system diseases. CNS Neuroscience & Therapeutics, 25(9), 899910. doi:10.1111/cns.13190CrossRefGoogle ScholarPubMed
Lopizzo, N., Bocchio Chiavetto, L., Cattane, N., Plazzotta, G., Tarazi, F. I., Pariante, C. M., … Cattaneo, A. (2015). Gene-environment interaction in major depression: Focus on experience-dependent biological systems. Frontiers in Psychiatry, 6, 68. doi:10.3389/fpsyt.2015.00068CrossRefGoogle ScholarPubMed
Mahady, L., Nadeem, M., Malek-Ahmadi, M., Chen, K., Perez, S. E., & Mufson, E. J. (2018). Frontal cortex epigenetic dysregulation during the progression of Alzheimer's disease. Journal of Alzheimer's Disease, 62(1), 115131. doi:10.3233/JAD-171032CrossRefGoogle ScholarPubMed
Malhi, G. S., & Mann, J. J. (2018). Depression. Lancet (London, England), 392(10161), 22992312. doi:10.1016/s0140-6736(18)31948-2CrossRefGoogle ScholarPubMed
Martin, T. C., Yet, I., Tsai, P. C., & Bell, J. T. (2015). coMET: Visualisation of regional epigenome-wide association scan results and DNA co-methylation patterns. BMC Bioinformatics, 16(1), 131. doi:10.1186/s12859-015-0568-2CrossRefGoogle ScholarPubMed
Miles, A. E., Dos Santos, F. C., Byrne, E. M., Renteria, M. E., McIntosh, A. M., Adams, M. J., … Nikolova, Y. S. (2021). Transcriptome-based polygenic score links depression-related corticolimbic gene expression changes to sex-specific brain morphology and depression risk. Neuropsychopharmacology, 46(13), 23042311. doi:10.1038/s41386-021-01189-xCrossRefGoogle ScholarPubMed
Nagel, M., Jansen, P. R., Stringer, S., Watanabe, K., de Leeuw, C. A., Bryois, J., … Posthuma, D. (2018). Meta-analysis of genome-wide association studies for neuroticism in 449484 individuals identifies novel genetic loci and pathways. Nature Genetics, 50(7), 920927. doi:10.1038/s41588-018-0151-7CrossRefGoogle Scholar
Nordlund, J., Bäcklin, C. L., Wahlberg, P., Busche, S., Berglund, E. C., Eloranta, M.-L., … Syvänen, A.-C. (2013). Genome-wide signatures of differential DNA methylation in pediatric acute lymphoblastic leukemia. Genome Biology, 14(9), r105. doi:10.1186/gb-2013-14-9-r105CrossRefGoogle ScholarPubMed
Oldfield, R. C. (1971). The assessment and analysis of handedness: The Edinburgh inventory. Neuropsychologia, 9(1), 97113. doi:10.1016/0028-3932(71)90067-4CrossRefGoogle ScholarPubMed
Olshen, A. B., Venkatraman, E. S., Lucito, R., & Wigler, M. (2004). Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics (Oxford, England), 5(4), 557572. doi:10.1093/biostatistics/kxh008CrossRefGoogle ScholarPubMed
Osimo, E. F., Pillinger, T., Rodriguez, I. M., Khandaker, G. M., Pariante, C. M., & Howes, O. D. (2020). Inflammatory markers in depression: A meta-analysis of mean differences and variability in 5166 patients and 5083 controls. Brain, Behavior, and Immunity, 87, 901909. doi:10.1016/j.bbi.2020.02.010CrossRefGoogle Scholar
Panizzon, M. S., Fennema-Notestine, C., Eyler, L. T., Jernigan, T. L., Prom-Wormley, E., Neale, M., … Kremen, W. S. (2009). Distinct genetic influences on cortical surface area and cortical thickness. Cerebral Cortex, 19(11), 27282735. doi:10.1093/cercor/bhp026CrossRefGoogle ScholarPubMed
Park, S.-Y., & Kim, J.-S. (2020). A short guide to histone deacetylases including recent progress on class II enzymes. Experimental & Molecular Medicine, 52(2), 204212. doi:10.1038/s12276-020-0382-4CrossRefGoogle Scholar
Penner-Goeke, S., & Binder, E. B. (2019). Epigenetics and depression. Dialogues in Clinical Neuroscience, 21(4), 397405. doi:10.31887/DCNS.2019.21.4/ebinderCrossRefGoogle ScholarPubMed
Phillips, M. L., Chase, H. W., Sheline, Y. I., Etkin, A., Almeida, J. R., Deckersbach, T., & Trivedi, M. H. (2015). Identifying predictors, moderators, and mediators of antidepressant response in major depressive disorder: Neuroimaging approaches. American Journal of Psychiatry, 172(2), 124138. doi:10.1176/appi.ajp.2014.14010076CrossRefGoogle ScholarPubMed
Pierce, A. M., & Keating, A. K. (2014). TAM Receptor tyrosine kinases: Expression, disease and oncogenesis in the central nervous system. Brain Research, 1542, 206. doi:10.1016/j.brainres.2013.10.049CrossRefGoogle ScholarPubMed
Puvogel, S., Alsema, A., Kracht, L., Webster, M. J., Weickert, C. S., Sommer, I. E. C., & Eggen, B. J. L. (2022). Single-nucleus RNA sequencing of midbrain blood-brain barrier cells in schizophrenia reveals subtle transcriptional changes with overall preservation of cellular proportions and phenotypes. Molecular Psychiatry, 27(11), 47314740. doi:10.1038/s41380-022-01796-0CrossRefGoogle ScholarPubMed
Richter, M., Murtaza, N., Scharrenberg, R., White, S. H., Johanns, O., Walker, S., … Calderon de Anda, F. (2019). Altered TAOK2 activity causes autism-related neurodevelopmental and cognitive abnormalities through RhoA signaling. Molecular Psychiatry, 24(9), 13291350. doi:10.1038/s41380-018-0025-5CrossRefGoogle ScholarPubMed
Rijlaarsdam, J., Cosin-Tomas, M., Schellhas, L., Abrishamcar, S., Malmberg, A., Neumann, A., … Cecil, C. A. M. (2023). DNA methylation and general psychopathology in childhood: An epigenome-wide meta-analysis from the PACE consortium. Molecular Psychiatry, 28(3), 11281136. doi:10.1038/s41380-022-01871-6CrossRefGoogle ScholarPubMed
Ritchie, M. E., Phipson, B., Wu, D., Hu, Y., Law, C. W., Shi, W., & Smyth, G. K. (2015). Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research, 43(7), e47. doi:10.1093/nar/gkv007CrossRefGoogle ScholarPubMed
Rive, M. M., van Rooijen, G., Veltman, D. J., Phillips, M. L., Schene, A. H., & Ruhé, H. G. (2013). Neural correlates of dysfunctional emotion regulation in major depressive disorder. A systematic review of neuroimaging studies. Neuroscience & Biobehavioral Reviews, 37(10 Pt 2), 25292553. doi:10.1016/j.neubiorev.2013.07.018CrossRefGoogle ScholarPubMed
Rutten, B. P. F., Vermetten, E., Vinkers, C. H., Ursini, G., Daskalakis, N. P., Pishva, E., … Boks, M. P. M. (2018). Longitudinal analyses of the DNA methylome in deployed military servicemen identify susceptibility loci for post-traumatic stress disorder. Molecular Psychiatry, 23(5), 11451156. doi:10.1038/mp.2017.120CrossRefGoogle ScholarPubMed
Sakamoto, K., Tamamura, Y., Katsube, K., & Yamaguchi, A. (2008). Zfp64 participates in Notch signaling and regulates differentiation in mesenchymal cells. Journal of Cell Science, 121(Pt 10), 16131623. doi:10.1242/jcs.023119CrossRefGoogle ScholarPubMed
Saxonov, S., Berg, P., & Brutlag, D. L. (2006). A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proceedings of the National Academy of Sciences, 103(5), 14121417. doi:10.1073/pnas.0510310103CrossRefGoogle ScholarPubMed
Schmaal, L., Hibar, D. P., Sämann, P. G., Hall, G. B., Baune, B. T., Jahanshad, N., … Veltman, D. J. (2017). Cortical abnormalities in adults and adolescents with major depression based on brain scans from 20 cohorts worldwide in the ENIGMA Major Depressive Disorder Working Group. Molecular Psychiatry, 22(6), 900909. doi:10.1038/mp.2016.60CrossRefGoogle ScholarPubMed
Schneider, I., Kugel, H., Redlich, R., Grotegerd, D., Bürger, C., Bürkner, P. C., … Hohoff, C. (2018). Association of serotonin transporter gene AluJb methylation with major depression, amygdala responsiveness, 5-HTTLPR/rs25531 polymorphism, and stress. Neuropsychopharmacology, 43(6), 13081316. doi:10.1038/npp.2017.273CrossRefGoogle ScholarPubMed
Ségonne, F., Pacheco, J., & Fischl, B. (2007). Geometrically accurate topology-correction of cortical surfaces using nonseparating loops. IEEE Transactions on Medical Imaging, 26(4), 518529. doi:10.1109/tmi.2006.887364CrossRefGoogle ScholarPubMed
Shaw, C. A., Li, Y., Wiszniewska, J., Chasse, S., Zaidi, S. N. Y., Jin, W., … Szigeti, K. (2011). Olfactory copy number association with age at onset of Alzheimer disease. Neurology, 76(15), 13021309. doi:10.1212/WNL.0b013e3182166df5CrossRefGoogle ScholarPubMed
Sherva, R., Zhang, R., Sahelijo, N., Jun, G., Anglin, T., Chanfreau, C., … Logue, M. W. (2023). African ancestry GWAS of dementia in a large military cohort identifies significant risk loci. Molecular Psychiatry, 28(3), 12931302. doi:10.1038/s41380-022-01890-3CrossRefGoogle Scholar
Shinde, V., Sobreira, N., Wohler, E. S., Maiti, G., Hu, N., Silvestri, G., … Chakravarti, S. (2021). Pathogenic alleles in microtubule, secretory granule and extracellular matrix-related genes in familial keratoconus. Human Molecular Genetics, 30(8), 658671. doi:10.1093/hmg/ddab075CrossRefGoogle ScholarPubMed
Stein, J. L., Medland, S. E., Vasquez, A. A., Hibar, D. P., Senstad, R. E., Winkler, A. M., … Thompson, P. M. (2012). Identification of common variants associated with human hippocampal and intracranial volumes. Nature Genetics, 44(5), 552561. doi:10.1038/ng.2250CrossRefGoogle ScholarPubMed
Steinberg, S., de Jong, S., Mattheisen, M., Costas, J., Demontis, D., Jamain, S., … Stefansson, K. (2014). Common variant at 16p11.2 conferring risk of psychosis. Molecular Psychiatry, 19(1), 108114. doi:10.1038/mp.2012.157CrossRefGoogle ScholarPubMed
Sudarov, A., Zhang, X. J., Braunstein, L., LoCastro, E., Singh, S., Taniguchi, Y., … Ross, M. E. (2018). Mature hippocampal neurons require LIS1 for synaptic integrity: Implications for cognition. Biological Psychiatry, 83(6), 518529. doi:10.1016/j.biopsych.2017.09.011CrossRefGoogle ScholarPubMed
Suderman, M., Staley, J. R., French, R., Arathimos, R., Simpkin, A., & Tilling, K. (2018). Dmrff: Identifying differentially methylated regions efficiently with power and control. BioRxiv, 508556. doi:10.1101/508556Google Scholar
Suh, J. S., Schneider, M. A., Minuzzi, L., MacQueen, G. M., Strother, S. C., Kennedy, S. H., & Frey, B. N. (2019). Cortical thickness in major depressive disorder: A systematic review and meta-analysis. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 88, 287302. doi:10.1016/j.pnpbp.2018.08.008CrossRefGoogle ScholarPubMed
Tabarés-Seisdedos, R., Escámez, T., Martínez-Giménez, J. A., Balanzá, V., Salazar, J., Selva, G., … Martínez, S. (2006). Variations in genes regulating neuronal migration predict reduced prefrontal cognition in schizophrenia and bipolar subjects from Mediterranean Spain: A preliminary study. Neuroscience, 139(4), 12891300. doi:10.1016/j.neuroscience.2006.01.054CrossRefGoogle ScholarPubMed
Teschendorff, A. E., Marabita, F., Lechner, M., Bartlett, T., Tegner, J., Gomez-Cabrero, D., & Beck, S. (2013). A beta-mixture quantile normalization method for correcting probe design bias in illumina infinium 450 k DNA methylation data. Bioinformatics (Oxford, England), 29(2), 189196. doi:10.1093/bioinformatics/bts680Google ScholarPubMed
Tian, Y., Morris, T. J., Webster, A. P., Yang, Z., Beck, S., Feber, A., & Teschendorff, A. E. (2017). ChAMP: Updated methylation analysis pipeline for Illumina BeadChips. Bioinformatics (Oxford, England), 33(24), 39823984. doi:10.1093/bioinformatics/btx513Google ScholarPubMed
Tozzi, L., Farrell, C., Booij, L., Doolin, K., Nemoda, Z., Szyf, M., … Frodl, T. (2018). Epigenetic changes of FKBP5 as a link connecting genetic and environmental risk factors with structural and functional brain changes in major depression. Neuropsychopharmacology, 43(5), 11381145. doi:10.1038/npp.2017.290CrossRefGoogle ScholarPubMed
Troubat, R., Barone, P., Leman, S., Desmidt, T., Cressant, A., Atanasova, B., … Belzung, C. (2021). Neuroinflammation and depression: A review. European Journal of Neuroscience, 53(1), 151171. doi:10.1111/ejn.14720CrossRefGoogle ScholarPubMed
Uchida, S., Yamagata, H., Seki, T., & Watanabe, Y. (2018). Epigenetic mechanisms of major depression: Targeting neuronal plasticity. Psychiatry and Clinical Neurosciences, 72(4), 212227. doi:10.1111/pcn.12621CrossRefGoogle ScholarPubMed
Um, J. W., Pramanik, G., Ko, J. S., Song, M. Y., Lee, D., Kim, H., … Ko, J. (2014). Calsyntenins function as synaptogenic adhesion molecules in concert with neurexins. Cell Reports, 6(6), 10961109. doi:10.1016/j.celrep.2014.02.010CrossRefGoogle ScholarPubMed
Van der Auwera, S., Peyrot, W. J., Milaneschi, Y., Hertel, J., Baune, B., Breen, G., … Grabe, H. (2018). Genome-wide gene-environment interaction in depression: A systematic evaluation of candidate genes: The childhood trauma working-group of PGC-MDD. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics, 177(1), 4049. doi:10.1002/ajmg.b.32593CrossRefGoogle ScholarPubMed
Wheater, E. N. W., Stoye, D. Q., Cox, S. R., Wardlaw, J. M., Drake, A. J., Bastin, M. E., & Boardman, J. P. (2020). DNA methylation and brain structure and function across the life course: A systematic review. Neuroscience & Biobehavioral Reviews, 113, 133156. doi:10.1016/j.neubiorev.2020.03.007CrossRefGoogle ScholarPubMed
Winham, S. J., Cuellar-Barboza, A. B., McElroy, S. L., Oliveros, A., Crow, S., Colby, C. L., … Biernacka, J. M. (2014). Bipolar disorder with comorbid binge eating history: A genome-wide association study implicates APOB. Journal of Affective Disorders, 165, 151158. doi:10.1016/j.jad.2014.04.026CrossRefGoogle ScholarPubMed
Won, E., Choi, S., Kang, J., Kim, A., Han, K. M., Chang, H. S., … Ham, B. J. (2016). Association between reduced white matter integrity in the corpus callosum and serotonin transporter gene DNA methylation in medication-naive patients with major depressive disorder. Translational Psychiatry, 6(8), e866. doi:10.1038/tp.2016.137CrossRefGoogle ScholarPubMed
Woodbury-Smith, M., Lamoureux, S., Begum, G., Nassir, N., Akter, H., O'Rielly, D. D., … Uddin, M. (2022). Mutational landscape of autism spectrum disorder brain tissue. Genes, 13(2), 207. doi:10.3390/genes13020207CrossRefGoogle ScholarPubMed
Wu, H., Li, H., Bai, T., Han, L., Ou, J., Xun, G., … Xia, K. (2020). Phenotype-to-genotype approach reveals head-circumference-associated genes in an autism spectrum disorder cohort. Clinical Genetics, 97(2), 338346. doi:10.1111/cge.13665CrossRefGoogle Scholar
Xu, C., Mullersman, J. E., Wang, L., Bin Su, B., Mao, C., Posada, Y., … Wang, K.-S. (2013). Polymorphisms in seizure 6-like gene are associated with bipolar disorder I: Evidence of gene × gender interaction. Journal of Affective Disorders, 145(1), 9599. doi:10.1016/j.jad.2012.07.017CrossRefGoogle ScholarPubMed
Xue, W., Tan, W., Dong, L., Tang, Q., Yang, F., Shi, X., … Qian, Y. (2020). TNFAIP8 influences the motor function in mice after spinal cord injury (SCI) through meditating inflammation dependent on AKT. Biochemical and Biophysical Research Communications, 528(1), 234241. doi:10.1016/j.bbrc.2020.05.029CrossRefGoogle ScholarPubMed
Yrondi, A., Fiori, L. M., Nogovitsyn, N., Hassel, S., Théroux, J. F., Aouabed, Z., … Turecki, G. (2021). Association between the expression of lncRNA BASP-AS1 and volume of right hippocampal tail moderated by episode duration in major depressive disorder: A CAN-BIND 1 report. Translational Psychiatry, 11(1), 469. doi:10.1038/s41398-021-01592-4CrossRefGoogle ScholarPubMed
Zhang, H. F., Mellor, D., & Peng, D. H. (2018). Neuroimaging genomic studies in major depressive disorder: A systematic review. CNS Neuroscience & Therapeutics, 24(11), 10201036. doi:10.1111/cns.12829CrossRefGoogle ScholarPubMed
Zhang, Y., Su, Q., Xia, W., Jia, K., Meng, D., Wang, X., … Su, Z. (2023). MiR-140-3p directly targets Tyro3 to regulate OGD/R-induced neuronal injury through the PI3K/Akt pathway. Brain Research Bulletin, 192, 93106. doi:10.1016/j.brainresbull.2022.11.007CrossRefGoogle ScholarPubMed
Zhou, W., Chen, L., Jiang, B., Sun, Y., Li, M., Wu, H., … Qin, S. (2021). Large-scale whole-exome sequencing association study identifies FOXH1 gene and sphingolipid metabolism pathway influencing major depressive disorder. CNS Neuroscience & Therapeutics, 27(11), 14251428. doi:10.1111/cns.13733CrossRefGoogle ScholarPubMed
Zhou, W., Laird, P. W., & Shen, H. (2017). Comprehensive characterization, annotation and innovative use of Infinium DNA methylation BeadChip probes. Nucleic Acids Research, 45(4), e22. doi:10.1093/nar/gkw967Google ScholarPubMed
Supplementary material: File

Yang et al. supplementary material 1

Yang et al. supplementary material
Download Yang et al. supplementary material 1(File)
File 4.1 MB
Supplementary material: File

Yang et al. supplementary material 2

Yang et al. supplementary material
Download Yang et al. supplementary material 2(File)
File 312.6 KB