Circulating microRNA associated with future relapse status in major depressive disorder

Qingqin S Li, David Galbraith, Randall L Morrison, Madhukar H Trivedi, Wayne C Drevets, Qingqin S Li, David Galbraith, Randall L Morrison, Madhukar H Trivedi, Wayne C Drevets

Abstract

Major depressive disorder (MDD) is an episodic condition with relapsing and remitting disease course. Elucidating biomarkers that can predict future relapse in individuals responding to an antidepressant treatment holds the potential to identify those patients who are prone to illness recurrence. The current study explored relationships between relapse risk in recurrent MDD and circulating microRNAs (miRNAs) that participate in RNA silencing and post-transcriptional regulation of gene expression. Serum samples were acquired from individuals with a history of recurrent MDD who were followed longitudinally in the observational study, OBSERVEMDD0001 (ClinicalTrials.gov Identifier: NCT02489305). Circulating miRNA data were obtained in 63 participants who relapsed ("relapsers") and 154 participants who did not relapse ("non-relapsers") during follow-up. The miRNA was quantified using the ID3EAL™ miRNA Discovery Platform from MiRXES measuring 575 circulating miRNAs using a patented qPCR technology and normalized with a standard curve from spike-in controls in each plate. The association between miRNAs and subsequent relapse was tested using a linear model, adjusting for age, gender, and plate. Four miRNAs were nominally associated with relapse status during the observational follow-up phase with a false discover rate adjusted p-value < 0.1. Enrichment analysis of experimentally validated targets revealed 112 significantly enriched pathways, including neurogenesis, response to cytokine, neurotrophin signaling, vascular endothelial growth factor signaling, relaxin signaling, and cellular senescence pathways. These data suggest these miRNAs putatively associated with relapse status may have the potential to regulate genes involved in multiple signaling pathways that have previously been associated with MDD. If shown to be significant in a larger, independent sample, these data may hold potential for developing a miRNA signature to identify patients likely to relapse, allowing for earlier intervention.

Keywords: circulating miRNA; depression; hsa-miR-199b-5p; hsa-miR-215-5p; relapse.

Conflict of interest statement

QL and WD are employees of Janssen Research & Development, LLC, of Johnson & Johnson, and may hold equity in Johnson & Johnson. RM is an employee of Janssen Research & Development, LLC when the study was conducted; he is now retired and a consultant to Janssen Research & Development, LLC. MT is or has been an advisor/consultant and received fee from (lifetime disclosure): Abbott Laboratories, Inc., Abdi Ibrahim, Akzo (Organon Pharmaceuticals Inc.), Alkermes, AstraZeneca, Axon Advisors, Bristol-Myers Squibb Company, Cephalon, Inc., Cerecor, CME Institute of Physicians, Concert Pharmaceuticals, Inc., Eli Lilly and Company, Evotec, Fabre Kramer Pharmaceuticals, Inc., Forest Pharmaceuticals, GlaxoSmithKline, Janssen Global Services, LLC, Janssen Pharmaceutica Products, LP, Johnson & Johnson PRD, Libby, Lundbeck, Meade Johnson, MedAvante, Medtronic, Merck, Mitsubishi Tanabe Pharma Development America, Inc., Naurex, Neuronetics, Otsuka Pharmaceuticals, Pamlab, Parke-Davis Pharmaceuticals, Inc., Pfizer Inc., PgxHealth, Phoenix Marketing Solutions, Rexahn Pharmaceuticals, Ridge Diagnostics, Roche Products Ltd., Sepracor, SHIRE Development, Sierra, SK Life and Science, Sunovion, Takeda, Tal Medical/Puretech Venture, Targacept, Transcept, VantagePoint, Vivus, and Wyeth-Ayerst Laboratories. The remaining author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Li, Galbraith, Morrison, Trivedi and Drevets.

Figures

FIGURE 1
FIGURE 1
Volcano plot. logFC for relapser vs. non-relapser was plotted against -logP, where P is the association p-value.
FIGURE 2
FIGURE 2
Violin plots for the differentially expressed miRNA (FDR

FIGURE 3

Kaplan–Meier curves for (A) hsa-miR-200a-3p…

FIGURE 3

Kaplan–Meier curves for (A) hsa-miR-200a-3p , (B) hsa-miR-215-5p .

FIGURE 3
Kaplan–Meier curves for (A)hsa-miR-200a-3p, (B)hsa-miR-215-5p.

FIGURE 4

KEGG gene set enrichment analysis…

FIGURE 4

KEGG gene set enrichment analysis results. Targets of miRNA were plotted against the…

FIGURE 4
KEGG gene set enrichment analysis results. Targets of miRNA were plotted against the enriched pathway (A). Selected enriched gene sets were also plotted with p-value (B) and gene ratio (C) on x-axis.
FIGURE 3
FIGURE 3
Kaplan–Meier curves for (A)hsa-miR-200a-3p, (B)hsa-miR-215-5p.
FIGURE 4
FIGURE 4
KEGG gene set enrichment analysis results. Targets of miRNA were plotted against the enriched pathway (A). Selected enriched gene sets were also plotted with p-value (B) and gene ratio (C) on x-axis.

References

    1. Price JL, Drevets WC. Neural circuits underlying the pathophysiology of mood disorders. Trends Cogn Sci. (2012) 16:61–71.
    1. Drevets WC, Wittenberg GM, Bullmore ET, Manji HK. Immune targets for therapeutic development in depression: towards precision medicine. Nat Rev Drug Discov. (2022) 21:224–44. 10.1038/s41573-021-00368-1
    1. Kang SG, Cho SE. Neuroimaging biomarkers for predicting treatment response and recurrence of major depressive disorder. Int J Mol Sci. (2020) 21:2148.
    1. Ronold EH, Joormann J, Hammar A. Facing recovery: emotional bias in working memory, rumination, relapse, and recurrence of major depression; an experimental paradigm conducted five years after first episode of major depression. Appl Neuropsychol Adult. (2020) 27:299–310. 10.1080/23279095.2018.1550406
    1. Kennis M, Gerritsen L, Van Dalen M, Williams A, Cuijpers P, Bockting C. Prospective biomarkers of major depressive disorder: a systematic review and meta-analysis. Mol Psychiatry. (2020) 25:321–38.
    1. Rush AJ. Developing the evidence for evidence-based practice. CMAJ. (2008) 178:1313–5.
    1. Wray NR, Ripke S, Mattheisen M, Trzaskowski M, Byrne EM, Abdellaoui A, et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat Genet. (2018) 50:668–81. 10.1038/s41588-018-0090-3
    1. Howard DM, Adams MJ, Clarke TK, Hafferty JD, Gibson J, Shirali M, et al. Genome-wide meta-analysis of depression identifies 102 independent variants and highlights the importance of the prefrontal brain regions. Nat Neurosci. (2019) 22:343–52. 10.1038/s41593-018-0326-7
    1. Zhao MZ, Song XS, Ma JS. Gene x environment interaction in major depressive disorder. World J Clin Cases. (2021) 9:9368–75. 10.12998/wjcc.v9.i31.9368
    1. Bockmuhl Y, Patchev AV, Madejska A, Hoffmann A, Sousa JC, Sousa N, et al. Methylation at the CpG island shore region upregulates Nr3c1 promoter activity after early-life stress. Epigenetics. (2015) 10:247–57. 10.1080/15592294.2015.1017199
    1. Van Bodegom M, Homberg JR, Henckens M. Modulation of the hypothalamic-pituitary-adrenal axis by early life stress exposure. Front Cell Neurosci. (2017) 11:87. 10.3389/fncel.2017.00087
    1. Silva RC, Maffioletti E, Gennarelli M, Baune BT, Minelli A. Biological correlates of early life stressful events in major depressive disorder. Psychoneuroendocrinology. (2021) 125:105103.
    1. Dwivedi Y. Emerging role of microRNAs in major depressive disorder: diagnosis and therapeutic implications. Dialogues Clin Neurosci. (2014) 16:43–61.
    1. Kichukova TM, Popov NT, Ivanov HY, Vachev TI. Circulating microRNAs as a novel class of potential diagnostic biomarkers in neuropsychiatric disorders. Folia Med. (2015) 57:159–72. 10.1515/folmed-2015-0035
    1. Dwivedi Y. Pathogenetic and therapeutic applications of microRNAs in major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry. (2016) 64:341–8. 10.1016/j.pnpbp.2015.02.003
    1. Yuan H, Mischoulon D, Fava M, Otto MW. Circulating microRNAs as biomarkers for depression: many candidates, few finalists. J Affect Disord. (2018) 233:68–78. 10.1016/j.jad.2017.06.058
    1. Roy B, Yoshino Y, Allen L, Prall K, Schell G, Dwivedi Y. Exploiting Circulating MicroRNAs as Biomarkers in Psychiatric Disorders. Mol Diagn Ther. (2020) 24:279–98.
    1. Miao C, Chang J. The important roles of microRNAs in depression: new research progress and future prospects. J Mol Med. (2021) 99:619–36. 10.1007/s00109-021-02052-8
    1. Mohr AM, Mott JL. Overview of microRNA biology. Semin Liver Dis. (2015) 35:3–11.
    1. Gui Y, Liu H, Zhang L, Lv W, Hu X. Altered microRNA profiles in cerebrospinal fluid exosome in Parkinson disease and Alzheimer disease. Oncotarget. (2015) 6:37043–53. 10.18632/oncotarget.6158
    1. Lu TX, Rothenberg ME. MicroRNA. J Allergy Clin Immunol. (2018) 141:1202–7.
    1. Kourembanas S. Exosomes: vehicles of intercellular signaling, biomarkers, and vectors of cell therapy. Annu Rev Physiol. (2015) 77:13–27. 10.1146/annurev-physiol-021014-071641
    1. Simeone P, Bologna G, Lanuti P, Pierdomenico L, Guagnano MT, Pieragostino D, et al. Extracellular vesicles as signaling mediators and disease biomarkers across biological barriers. Int J Mol Sci. (2020) 21:2514.
    1. Szilagyi M, Pos O, Marton E, Buglyo G, Soltesz B, Keseru J, et al. Circulating Cell-Free Nucleic Acids: main Characteristics and Clinical Application. Int J Mol Sci. (2020) 21:6827.
    1. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. (2008) 105:10513–8.
    1. Wang X, Sundquist J, Zoller B, Memon AA, Palmer K, Sundquist K, et al. Determination of 14 circulating microRNAs in Swedes and Iraqis with and without diabetes mellitus type 2. PLoS One. (2014) 9:e86792. 10.1371/journal.pone.0086792
    1. Bye A, Rosjo H, Nauman J, Silva GJ, Follestad T, Omland T, et al. Circulating microRNAs predict future fatal myocardial infarction in healthy individuals – The HUNT study. J Mol Cell Cardiol. (2016) 97:162–8. 10.1016/j.yjmcc.2016.05.009
    1. Salloum-Asfar S, Satheesh NJ, Abdulla SA. Circulating miRNAs, small but promising biomarkers for autism spectrum disorder. Front Mol Neurosci. (2019) 12:253. 10.3389/fnmol.2019.00253
    1. Camkurt MA, Acar S, Coskun S, Gunes M, Gunes S, Yilmaz MF, et al. Comparison of plasma MicroRNA levels in drug naive, first episode depressed patients and healthy controls. J Psychiatr Res. (2015) 69:67–71.
    1. Sun N, Lei L, Wang Y, Yang C, Liu Z, Li X, et al. Preliminary comparison of plasma notch-associated microRNA-34b and -34c levels in drug naive, first episode depressed patients and healthy controls. J Affect Disord. (2016) 194:109–14. 10.1016/j.jad.2016.01.017
    1. Gecys D, Dambrauskiene K, Simonyte S, Patamsyte V, Vilkeviciute A, Musneckis A, et al. Circulating hsa-let-7e-5p and hsa-miR-125a-5p as possible biomarkers in the diagnosis of major depression and bipolar disorders. Dis Markers. (2022) 2022:3004338. 10.1155/2022/3004338
    1. Wang X, Sundquist K, Hedelius A, Palmer K, Memon AA, Sundquist J. Circulating microRNA-144-5p is associated with depressive disorders. Clin Epigenet. (2015) 7:69. 10.1186/s13148-015-0099-8
    1. Zhang HP, Liu XL, Chen JJ, Cheng K, Bai SJ, Zheng P, et al. Circulating microRNA 134 sheds light on the diagnosis of major depressive disorder. Transl Psychiatry. (2020) 10:95. 10.1038/s41398-020-0773-2
    1. Roumans S, Sundquist K, Memon AA, Hedelius A, Sundquist J, Wang X. Association of circulating let-7b-5p with major depressive disorder: a nested case-control study. BMC Psychiatry. (2021) 21:616. 10.1186/s12888-021-03621-4
    1. Smalheiser NR, Lugli G, Zhang H, Rizavi H, Cook EH, Dwivedi Y. Expression of microRNAs and other small RNAs in prefrontal cortex in schizophrenia, bipolar disorder and depressed subjects. PLoS One. (2014) 9:e86469. 10.1371/journal.pone.0086469
    1. Gururajan A, Naughton ME, Scott KA, O’connor RM, Moloney G, Clarke G, et al. MicroRNAs as biomarkers for major depression: a role for let-7b and let-7c. Transl Psychiatry. (2016) 6:e862. 10.1038/tp.2016.131
    1. Liu W, Zhang F, Zheng Y, He S, Zhang T, Guo Q, et al. The role of circulating blood microRNA-374 and microRNA-10 levels in the pathogenesis and therapeutic mechanisms of major depressive disorder. Neurosci Lett. (2021) 763:136184. 10.1016/j.neulet.2021.136184
    1. So JBY, Kapoor R, Zhu F, Koh C, Zhou L, Zou R, et al. Development and validation of a serum microRNA biomarker panel for detecting gastric cancer in a high-risk population. Gut. (2021) 70:829–37. 10.1136/gutjnl-2020-322065
    1. Hoffman GE, Schadt EE. variancePartition: interpreting drivers of variation in complex gene expression studies. BMC Bioinformat. (2016) 17:483. 10.1186/s12859-016-1323-z
    1. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. (2015) 43:e47. 10.1093/nar/gkv007
    1. Therneau TM, Grambsch PM. Modeling Survival Data: Extending the Cox Model. New York, NY: Springer; (2000).
    1. Therneau T. A Package for Survival Analysis in R. R package version 3.2-7. Vienna: R Core Team; (2020).
    1. Kassambara A, Kosinski M, Biecek P. survminer: Drawing Survival Curves using “ggplot2”. R package version 0.4.9. Vienna: R Core Team; (2021).
    1. Xiao F, Zuo Z, Cai G, Kang S, Gao X, Li T. miRecords: an integrated resource for microRNA-target interactions. Nucleic Acids Res. (2009) 37:D105–10. 10.1093/nar/gkn851
    1. Karagkouni D, Paraskevopoulou MD, Chatzopoulos S, Vlachos IS, Tastsoglou S, Kanellos I. DIANA-TarBase v8: a decade-long collection of experimentally supported miRNA-gene interactions. Nucleic Acids Res. (2018) 46:D239–45. 10.1093/nar/gkx1141
    1. Agarwal V, Bell GW, Nam JW, Bartel DP. Predicting effective microRNA target sites in mammalian mRNAs. Elife. (2015) 4:e05005.
    1. Boyle EI, Weng S, Gollub J, Jin H, Botstein D, Cherry JM, et al. GO::TermFinder–open source software for accessing Gene Ontology information and finding significantly enriched Gene Ontology terms associated with a list of genes. Bioinformatics. (2004) 20:3710–5. 10.1093/bioinformatics/bth456
    1. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. (2000) 28:27–30.
    1. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. (2012) 16:284–7. 10.1089/omi.2011.0118
    1. Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics. (2011) 27:1739–40. 10.1093/bioinformatics/btr260
    1. Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. (2017) 45:D353–61.
    1. Mellios N, Feldman DA, Sheridan SD, Ip JPK, Kwok S, Amoah SK, et al. MeCP2-regulated miRNAs control early human neurogenesis through differential effects on ERK and AKT signaling. Mol Psychiatry. (2018) 23:1051–65. 10.1038/mp.2017.86
    1. Ausio J. MeCP2 and the enigmatic organization of brain chromatin. Implications for depression and cocaine addiction. Clin Epigenet. (2016) 8:58. 10.1186/s13148-016-0214-5
    1. Mcgill BE, Bundle SF, Yaylaoglu MB, Carson JP, Thaller C, Zoghbi HY. Enhanced anxiety and stress-induced corticosterone release are associated with increased Crh expression in a mouse model of Rett syndrome. Proc Natl Acad Sci USA. (2006) 103:18267–72. 10.1073/pnas.0608702103
    1. Murgatroyd C, Patchev AV, Wu Y, Micale V, Bockmuhl Y, Fischer D, et al. Dynamic DNA methylation programs persistent adverse effects of early-life stress. Nat Neurosci. (2009) 12:1559–66.
    1. Wu Y, Patchev AV, Daniel G, Almeida OF, Spengler D. Early-life stress reduces DNA methylation of the Pomc gene in male mice. Endocrinology. (2014) 155:1751–62. 10.1210/en.2013-1868
    1. Zimmermann CA, Hoffmann A, Raabe F, Spengler D. Role of mecp2 in experience-dependent epigenetic programming. Genes. (2015) 6:60–86.
    1. Chen WG, Chang Q, Lin Y, Meissner A, West AE, Griffith EC, et al. Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2. Science. (2003) 302:885–9. 10.1126/science.1086446
    1. Zhou Z, Hong EJ, Cohen S, Zhao WN, Ho HY, Schmidt L, et al. Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation. Neuron. (2006) 52:255–69. 10.1016/j.neuron.2006.09.037
    1. Gabel HW, Kinde B, Stroud H, Gilbert CS, Harmin DA, Kastan NR, et al. Disruption of DNA-methylation-dependent long gene repression in Rett syndrome. Nature. (2015) 522:89–93. 10.1038/nature14319
    1. Hutchinson AN, Deng JV, Cohen S, West AE. Phosphorylation of MeCP2 at Ser421 contributes to chronic antidepressant action. J Neurosci. (2012) 32:14355–63. 10.1523/JNEUROSCI.2156-12.2012
    1. Su M, Hong J, Zhao Y, Liu S, Xue X. MeCP2 controls hippocampal brain-derived neurotrophic factor expression via homeostatic interactions with microRNA132 in rats with depression. Mol Med Rep. (2015) 12:5399–406. 10.3892/mmr.2015.4104
    1. Dionisie V, Ciobanu AM, Toma VA, Manea MC, Baldea I, Olteanu D, et al. Escitalopram targets oxidative stress, caspase-3, BDNF and MeCP2 in the hippocampus and frontal cortex of a rat model of depression induced by chronic unpredictable mild stress. Int J Mol Sci. (2021) 22:7483. 10.3390/ijms22147483
    1. Van Der Sijde F, Vietsch EE, Mustafa DAM, Li Y, Van Eijck CHJ. Serum miR-338-3p and miR-199b-5p are associated with the absolute neutrophil count in patients with resectable pancreatic cancer. Clin Chim Acta. (2020) 505:183–9. 10.1016/j.cca.2020.03.005
    1. Zhang Y, Zhang X, Zhao Z, Zheng Y, Xiao Z, Li F. Integrated bioinformatics analysis and validation revealed potential immune-regulatory miR-892b, miR-199b-5p and miR-582-5p as diagnostic biomarkers in active tuberculosis. Microb Pathog. (2019) 134:103563. 10.1016/j.micpath.2019.103563
    1. Yoshino Y, Roy B, Dwivedi Y. Differential and unique patterns of synaptic miRNA expression in dorsolateral prefrontal cortex of depressed subjects. Neuropsychopharmacology. (2021) 46:900–10. 10.1038/s41386-020-00861-y
    1. Satyanarayanan SK, Shih YH, Wen YR, Palani M, Lin YW, Su H, et al. miR-200a-3p modulates gene expression in comorbid pain and depression: molecular implication for central sensitization. Brain Behav Immun. (2019) 82:230–8. 10.1016/j.bbi.2019.08.190
    1. Yang Z, Wang J, Pan Z, Zhang Y. miR-143-3p regulates cell proliferation and apoptosis by targeting IGF1R and IGFBP5 and regulating the Ras/p38 MAPK signaling pathway in rheumatoid arthritis. Exp Ther Med. (2018) 15:3781–90. 10.3892/etm.2018.5907
    1. Sun C, Jia N, Li R, Zhang Z, Zhong Y, Han K. miR-143-3p inhibition promotes neuronal survival in an Alzheimer’s disease cell model by targeting neuregulin-1. Folia Neuropathol. (2020) 58:10–21. 10.5114/fn.2020.94002

Source: PubMed

3
Iratkozz fel