1,912
Views
0
CrossRef citations to date
0
Altmetric
Basic Research Article

DNA methylation changes in association with trauma-focused psychotherapy efficacy in treatment-resistant depression patients: a prospective longitudinal study

Cambios en la metilación del ADN en asociación con la eficacia de la psicoterapia centrada en trauma en pacientes con depresión resistentes al tratamiento: un estudio longitudinal prospectivo

, , , , , , , & ORCID Icon show all
Article: 2314913 | Received 29 Aug 2023, Accepted 30 Jan 2024, Published online: 16 Feb 2024

References

  • Arab, A. H., & Elhawary, N. A. (2015). Association between ANKK1 (rs1800497) and LTA (rs909253) Genetic Variants and Risk of Schizophrenia. BioMed Research International, 1. https://doi.org/10.1155/2015/821827
  • Arranz, M. J., Gallego-Fabrega, C., Martín-Blanco, A., Soler, J., Elices, M., Dominguez-Clavé, E., Salazar, J., Vega, D., Briones-Buixassa, L., & Pascual, J. C. (2021). A genome-wide methylation study reveals X chromosome and childhood trauma methylation alterations associated with borderline personality disorder. Translational Psychiatry, 11(1), 5. https://doi.org/10.1038/s41398-020-01139-z
  • Bifulco, A., Bernazzani, O., Moran, P. M., & Jacobs, C. (2005). The childhood experience of care and abuse questionnaire (CECA.Q): Validation in a community series. British Journal of Clinical Psychology, 44(4), 563–581. https://doi.org/10.1348/014466505X35344
  • Catts, V. S., & Weickert, C. S. (2012). Gene expression analysis implicates a death receptor pathway in schizophrenia pathology. PLoS One, 7(4), e35511. https://doi.org/10.1371/journal.pone.0035511
  • Cecil, C. A. M., Smith, R. G., Walton, E., Mill, J., McCrory, E. J., & Viding, E. (2016). Epigenetic signatures of childhood abuse and neglect: Implications for psychiatric vulnerability. Journal of Psychiatric Research, 83, 184–194. https://doi.org/10.1016/j.jpsychires.2016.09.010
  • Dall’Aglio, L., Muka, T., Cecil, C. A. M., Bramer, W. M., Verbiest, M. M. P. J., Nano, J., Hidalgo, A. C., Franco, O. H., & Tiemeier, H. (2018). The role of epigenetic modifications in neurodevelopmental disorders: A systematic review. Neuroscience & Biobehavioral Reviews, 94, 17–30. https://doi.org/10.1016/j.neubiorev.2018.07.011
  • Dodd, S., Bauer, M., Carvalho, A. F., Eyre, H., Fava, M., Kasper, S., Kennedy, S. H., Khoo, J.-P., Lopez Jaramillo, C., Malhi, G. S., McIntyre, R. S., Mitchell, P. B., Castro, A. M. P., Ratheesh, A., Severus, E., Suppes, T., Trivedi, M. H., Thase, M. E., Yatham, L. N., … Berk, M. (2021). A clinical approach to treatment resistance in depressed patients: What to do when the usual treatments don’t work well enough? The World Journal of Biological Psychiatry, 22(7), 483–494. https://doi.org/10.1080/15622975.2020.1851052
  • Drago, A., Giegling, I., Schäfer, M., Hartmann, A. M., Konte, B., Friedl, M., Serretti, A., & Rujescu, D. (2014). Genome-wide association study supports the role of the immunological system and of the neurodevelopmental processes in response to haloperidol treatment. Pharmacogenetics and Genomics, 24(6), 314–319. https://doi.org/10.1097/FPC.0000000000000052
  • Du, P., Zhang, X., Huang, C.-C., Jafari, N., Kibbe, W. A., Hou, L., & Lin, S. M. (2010). Comparison of Beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics, 11(1), 587. https://doi.org/10.1186/1471-2105-11-587
  • Flory, J. D., & Yehuda, R. (2015). Comorbidity between post-traumatic stress disorder and major depressive disorder: alternative explanations and treatment considerations. Dialogues in Clinical Neuroscience, 17(2), 141–150. https://doi.org/10.31887/DCNS.2015.17.2/jflory
  • Fortin, J.-P., Triche, T. J., & Hansen, K. D. (2017). Preprocessing, normalization and integration of the Illumina HumanMethylationEPIC array with minfi. Bioinformatics (Oxford, England), 33(4), 558–560. https://doi.org/10.1093/bioinformatics/btw691
  • Gandal, M. J., Zhang, P., Hadjimichael, E., Walker, R. L., Chen, C., Liu, S., Won, H., van Bakel, H., Varghese, M., Wang, Y., Shieh, A. W., Haney, J., Parhami, S., Belmont, J., Kim, M., Moran Losada, P., Khan, Z., Mleczko, J., Xia, Y., … Geschwind, D. H. (2018). Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science, 362(6420), https://doi.org/10.1126/science.aat8127
  • GBD 2016. Disease and injury incidence and prevalence collaborators. (2016). Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015. The Lancet, 388(10053), 1545–1602. https://doi.org/10.1016/S0140-6736(16)31678-6
  • Hasin, D. S., Sarvet, A. L., Meyers, J. L., Saha, T. D., Ruan, W. J., Stohl, M., & Grant, B. F. (2018). Epidemiology of Adult DSM-5 Major Depressive Disorder and Its Specifiers in the United States. JAMA Psychiatry, 75(4), 336–346. https://doi.org/10.1001/jamapsychiatry.2017.4602
  • Huang, G., He, X., & Wei, X.-L. (2018). lncRNA NEAT1 promotes cell proliferation and invasion by regulating miR–365/RGS20 in oral squamous cell carcinoma. Oncology Reports, 39(4), 1948–1956. https://doi.org/10.3892/or.2018.6283
  • Jaffe, D. H., Rive, B., & Denee, T. R. (2019). The humanistic and economic burden of treatment-resistant depression in Europe: a cross-sectional study. BMC Psychiatry, 19(1), 247. https://doi.org/10.1186/s12888-019-2222-4
  • Kautzky, A., Baldinger-Melich, P., Kranz, G. S., Vanicek, T., Souery, D., Montgomery, S., Mendlewicz, J., Zohar, J., Serretti, A., Lanzenberger, R., & Kasper, S. (2017). A New prediction model for evaluating treatment-resistant depression. The Journal of Clinical Psychiatry, 78(2), 215–222. https://doi.org/10.4088/JCP.15m10381
  • Keshavarz, F., Soltani, M., Mokhtarian, K., Beshkar, P., Majidi, J., Azadegan-Dehkordi, F., Anjomshoa, M., & Bagheri, N. (2022). Autoantibodies against central nervous system antigens and the serum levels of IL-32 in patients with schizophrenia. Neuroimmunomodulation, 29(4), 493–499. https://doi.org/10.1159/000526425
  • Kikuchi, M., Yamada, K., Toyota, T., Itokawa, M., Hattori, E., Yoshitsugu, K., Shimizu, H., & Yoshikawa, T. (2003). Two-step association analyses of the chromosome 18p11.2 region in schizophrenia detect a locus encompassing C18orf1. Molecular Psychiatry, 8(5), 467–469. https://doi.org/10.1038/sj.mp.4001280
  • Knoblich, N., Gundel, F., Brückmann, C., Becker-Sadzio, J., Frischholz, C., & Nieratschker, V. (2018). DNA methylation of APBA3 and MCF2 in borderline personality disorder: Potential biomarkers for response to psychotherapy. European Neuropsychopharmacology, 28(2), 252–263. https://doi.org/10.1016/j.euroneuro.2017.12.010
  • Kraus, C., Kadriu, B., Lanzenberger, R., Zarate, C. A., & Kasper, S. (2019). Prognosis and improved outcomes in major depression: a review. Translational Psychiatry, 9(1), 127. https://doi.org/10.1038/s41398-019-0460-3
  • Labonté, B., Suderman, M., Maussion, G., Navaro, L., Yerko, V., Mahar, I., Bureau, A., Mechawar, N., Szyf, M., Meaney, M. J., & Turecki, G. (2012). Genome-wide epigenetic regulation by early-life trauma. Archives of General Psychiatry, 69(7), 722–731. https://doi.org/10.1001/archgenpsychiatry.2011.2287
  • Lanz, T. A., Reinhart, V., Sheehan, M. J., Rizzo, S. J. S., Bove, S. E., James, L. C., Volfson, D., Lewis, D. A., & Kleiman, R. J. (2019). Postmortem transcriptional profiling reveals widespread increase in inflammation in schizophrenia: A comparison of prefrontal cortex, striatum, and hippocampus among matched tetrads of controls with subjects diagnosed with schizophrenia, bipolar or major. Translational Psychiatry, 9(1), 151. https://doi.org/10.1038/s41398-019-0492-8
  • Liew, C.-C., Ma, J., Tang, H.-C., Zheng, R., & Dempsey, A. A. (2006). The peripheral blood transcriptome dynamically reflects system wide biology: A potential diagnostic tool. Journal of Laboratory and Clinical Medicine, 147(3), 126–132. https://doi.org/10.1016/j.lab.2005.10.005
  • Maffioletti, E., Minelli, A., Tardito, D., & Gennarelli, M. (2020). Blues in the brain and beyond: Molecular bases of major depressive disorder and relative pharmacological and Non-pharmacological treatments. Genes, 11(9), https://doi.org/10.3390/genes11091089
  • Marinova, Z., Maercker, A., Küffer, A., Robinson, M. D., Wojdacz, T. K., Walitza, S., Grünblatt, E., & Burri, A. (2017). DNA methylation profiles of elderly individuals subjected to indentured childhood labor and trauma. BMC Medical Genetics, 18(1), 21. https://doi.org/10.1186/s12881-017-0370-2
  • Mehta, D., Klengel, T., Conneely, K. N., Smith, A. K., Altmann, A., Pace, T. W., Rex-Haffner, M., Loeschner, A., Gonik, M., Mercer, K. B., Bradley, B., Müller-Myhsok, B., Ressler, K. J., & Binder, E. B. (2013). Childhood maltreatment is associated with distinct genomic and epigenetic profiles in posttraumatic stress disorder. Proceedings of the National Academy of Sciences, 110(20), 8302–8307. https://doi.org/10.1073/pnas.1217750110
  • Minelli, A., Magri, C., Giacopuzzi, E., & Gennarelli, M. (2018). The effect of childhood trauma on blood transcriptome expression in major depressive disorder. Journal of Psychiatric Research, 104, 50–54. https://doi.org/10.1016/j.jpsychires.2018.06.014
  • Minelli, A., Zampieri, E., Sacco, C., Bazzanella, R., Mezzetti, N., Tessari, E., Barlati, S., & Bortolomasi, M. (2019). Clinical efficacy of trauma-focused psychotherapies in treatment-resistant depression (TRD) in-patients: A randomized, controlled pilot-study. Psychiatry Research, 273, 567–574. https://doi.org/10.1016/j.psychres.2019.01.070
  • Miyake, K., Miyashita, C., Ikeda-Araki, A., Miura, R., Itoh, S., Yamazaki, K., Kobayashi, S., Masuda, H., Ooka, T., Yamagata, Z., & Kishi, R. (2021). DNA methylation of GFI1 as a mediator of the association between prenatal smoking exposure and ADHD symptoms at 6 years: The Hokkaido Study on Environment and Children’s Health. Clinical Epigenetics, 13(1), 74. https://doi.org/10.1186/s13148-021-01063-z
  • Montgomery, S. A., & Asberg, M. (1979). A new depression scale designed to be sensitive to change. British Journal of Psychiatry, 134(4), 382–389. https://doi.org/10.1192/bjp.134.4.382
  • Moschny, N., Zindler, T., Jahn, K., Dorda, M., Davenport, C. F., Wiehlmann, L., Maier, H. B., Eberle, F., Bleich, S., Neyazi, A., & Frieling, H. (2020). Novel candidate genes for ECT response prediction-a pilot study analyzing the DNA methylome of depressed patients receiving electroconvulsive therapy. Clinical Epigenetics, 12(1), 114. https://doi.org/10.1186/s13148-020-00891-9
  • Müller, N., Krause, D., Barth, R., Myint, A.-M., Weidinger, E., Stettinger, W., Zill, P., Drexhage, H., & Schwarz, M. J. (2019). Childhood adversity and current stress are related to Pro- and anti-inflammatory cytokines in major depression. Journal of Affective Disorders, 253, 270–276. https://doi.org/10.1016/j.jad.2019.04.088
  • Neves, I., Dinis-Oliveira, R. J., & Magalhães, T. (2021). Epigenomic mediation after adverse childhood experiences: A systematic review and meta-analysis. Forensic Sciences Research, 6(2), 103–114. https://doi.org/10.1080/20961790.2019.1641954
  • O’Donnell, K. J., Chen, L., MacIsaac, J. L., McEwen, L. M., Nguyen, T., Beckmann, K., Zhu, Y., Chen, L. M., Brooks-Gunn, J., Goldman, D., Grigorenko, E. L., Leckman, J. F., Diorio, J., Karnani, N., Olds, D. L., Holbrook, J. D., Kobor, M. S., & Meaney, M. J. (2018). DNA methylome variation in a perinatal nurse-visitation program that reduces child maltreatment: A 27-year follow-up. Translational Psychiatry, 8(1), 15. https://doi.org/10.1038/s41398-017-0063-9
  • Pandey, G. N., Rizavi, H. S., Zhang, H., & Ren, X. (2018). Abnormal gene and protein expression of inflammatory cytokines in the postmortem brain of schizophrenia patients. Schizophrenia Research, 192, 247–254. https://doi.org/10.1016/j.schres.2017.04.043
  • Parade, S. H., Huffhines, L., Daniels, T. E., Stroud, L. R., Nugent, N. R., & Tyrka, A. R. (2021). A systematic review of childhood maltreatment and DNA methylation: Candidate gene and epigenome-wide approaches. Translational Psychiatry, 11(1), 134. https://doi.org/10.1038/s41398-021-01207-y
  • Pedicone, C., Meyer, S. T., Chisholm, J. D., & Kerr, W. G. (2021). Targeting SHIP1 and SHIP2 in Cancer. Cancers, 13(4)), https://doi.org/10.3390/cancers13040890
  • Perroud, N., Salzmann, A., Prada, P., Nicastro, R., Hoeppli, M. E., Furrer, S., Ardu, S., Krejci, I., Karege, F., & Malafosse, A. (2013). Response to psychotherapy in borderline personality disorder and methylation status of the BDNF gene. Translational Psychiatry, 3(1), e207. https://doi.org/10.1038/tp.2012.140
  • Pidsley, R., Zotenko, E., Peters, T. J., Lawrence, M. G., Risbridger, G. P., Molloy, P., Van Djik, S., Muhlhausler, B., Stirzaker, C., & Clark, S. J. (2016). Critical evaluation of the Illumina MethylationEPIC BeadChip microarray for whole-genome DNA methylation profiling. Genome Biology, 17(1), 208. https://doi.org/10.1186/s13059-016-1066-1
  • Sales, G., Calura, E., & Romualdi, C. (2019). metaGraphite-a new layer of pathway annotation to get metabolite networks. Bioinformatics (Oxford, England), 35(7), 1258–1260. https://doi.org/10.1093/bioinformatics/bty719
  • Silva, R. C., Maffioletti, E., Gennarelli, M., Baune, B. T., & Minelli, A. (2021). Biological correlates of early life stressful events in major depressive disorder. Psychoneuroendocrinology, 125, 105103. https://doi.org/10.1016/j.psyneuen.2020.105103
  • Sirignano, L., Frank, J., Kranaster, L., Witt, S. H., Streit, F., Zillich, L., Sartorius, A., Rietschel, M., & Foo, J. C. (2021). Methylome-wide change associated with response to electroconvulsive therapy in depressed patients. Translational Psychiatry, 11(1), 347. https://doi.org/10.1038/s41398-021-01474-9
  • Snijders, C., Maihofer, A. X., Ratanatharathorn, A., Baker, D. G., Boks, M. P., Geuze, E., Jain, S., Kessler, R. C., Pishva, E., Risbrough, V. B., Stein, M. B., Ursano, R. J., Vermetten, E., Vinkers, C. H., Smith, A. K., Uddin, M., Rutten, B. P. F., PGC PTSD EWAS Consortium, & Nievergelt, C. M. (2020). Longitudinal epigenome-wide association studies of three male military cohorts reveal multiple CpG sites associated with post-traumatic stress disorder. Clinical Epigenetics, 12(1), 11. https://doi.org/10.1186/s13148-019-0798-7
  • Thase, M. E., & Rush, A. J. (1997). When at first you don’t succeed: Sequential strategies for antidepressant nonresponders. The Journal of Clinical Psychiatry, 58(Suppl 1), 16–21. https://doi.org/10.4088/JCP.v58n0103
  • Van Assche, E., Hohoff, C., Zang, J., Knight, M. J., & Baune, B. T. (2023). Epigenetic modification related to cognitive changes during a cognitive training intervention in depression. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 127, 110835. https://doi.org/10.1016/j.pnpbp.2023.110835
  • Vinkers, C. H., Geuze, E., van Rooij, S. J. H., Kennis, M., Schür, R. R., Nispeling, D. M., Smith, A. K., Nievergelt, C. M., Uddin, M., Rutten, B. P. F., Vermetten, E., & Boks, M. P. (2021). Successful treatment of post-traumatic stress disorder reverses DNA methylation marks. Molecular Psychiatry, 26(4), 1264–1271. https://doi.org/10.1038/s41380-019-0549-3
  • Wang, P., Wang, H., Li, X., Liu, Y., Zhao, C., & Zhu, D. (2016). Srcin1 suppressed osteosarcoma cell proliferation and invasion. PLoS One, 11(8), e0155518. https://doi.org/10.1371/journal.pone.0155518
  • Wu, W., Howard, D., Sibille, E., & French, L. (2021). Differential and spatial expression meta-analysis of genes identified in genome-wide association studies of depression. Translational Psychiatry, 11(1), 8. https://doi.org/10.1038/s41398-020-01127-3
  • Yan, S., Shan, Y., Zhong, S., Miao, H., Luo, Y., Ran, H., & Jia, Y. (2021). The effectiveness of Eye movement desensitization and reprocessing toward adults with major depressive disorder: A meta-analysis of randomized controlled trials. Frontiers in Psychiatry, 12, 700458. https://doi.org/10.3389/fpsyt.2021.700458
  • Yang, R., Xu, C., Bierer, L. M., Flory, J. D., Gautam, A., Bader, H. N., Lehrner, A., Makotkine, I., Desarnaud, F., Miller, S. A., Jett, M., Hammamieh, R., & Yehuda, R. (2021). Longitudinal genome-wide methylation study of PTSD treatment using prolonged exposure and hydrocortisone. Translational Psychiatry, 11(1), 398. https://doi.org/10.1038/s41398-021-01513-5
  • Zhang, M., Ma, F., Xie, R., Wu, Y., Wu, M., Zhang, P., Peng, Y., Zhao, J., Xiong, J., Li, A., Kequan, C., Zhang, Y., Liu, S., Wang, J., & Chen, X. (2017). Overexpression of Srcin1 contributes to the growth and metastasis of colorectal cancer. International Journal of Oncology, 50(5), 1555–1566. https://doi.org/10.3892/ijo.2017.3952
  • Zhang, Z., Zheng, F., You, Y., Ma, Y., Lu, T., Yue, W., & Zhang, D. (2016). Growth arrest specific gene 7 is associated with schizophrenia and regulates neuronal migration and morphogenesis. Molecular Brain, 9(1), 54. https://doi.org/10.1186/s13041-016-0238-y
  • Zheng, Y., Lunetta, K. L., Liu, C., Katrinli, S., Smith, A. K., Miller, M. W., & Logue, M. W. (2022). An evaluation of the genome-wide false positive rates of common methods for identifying differentially methylated regions using illumina methylation arrays. Epigenetics, 17(13), 2241–2258. https://doi.org/10.1080/15592294.2022.2115600
  • Zhuang, H., Wang, H., & Ji, Z. (2022). findPC: An R package to automatically select the number of principal components in single-cell analysis. Bioinformatics (Oxford, England), 38(10), 2949–2951. https://doi.org/10.1093/bioinformatics/btac235
  • Ziegler, C., Richter, J., Mahr, M., Gajewska, A., Schiele, M. A., Gehrmann, A., Schmidt, B., Lesch, K.-P., Lang, T., Helbig-Lang, S., Pauli, P., Kircher, T., Reif, A., Rief, W., Vossbeck-Elsebusch, A. N., Arolt, V., Wittchen, H.-U., Hamm, A. O., Deckert, J., & Domschke, K. (2016). MAOA gene hypomethylation in panic disorder-reversibility of an epigenetic risk pattern by psychotherapy. Translational Psychiatry, 6(4), e773. https://doi.org/10.1038/tp.2016.41