Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Can Molecular Biology Propose Reliable Biomarkers for Diagnosing Major Depression?

Author(s): Nikolay N. Ivanets, Andrey A. Svistunov, Vladimir N. Chubarev*, Marina A. Kinkulkina, Yuliya G. Tikhonova, Nikita S. Syzrantsev, Susanna S. Sologova, Nelly V. Ignatyeva, Kerim Mutig and Vadim V. Tarasov

Volume 27, Issue 2, 2021

Published on: 24 November, 2020

Page: [305 - 318] Pages: 14

DOI: 10.2174/1381612826666201124110437

Price: $65

Abstract

Background: Modern medicine has provided considerable knowledge of the pathophysiology of mental disorders at the body, systemic, organ and neurochemical levels of the biological organization of the body. Modern clinical diagnostics of depression have some problems, that is why psychiatric society makes use of diagnostics and taxonomy of different types of depression by implemention of modern molecular biomarkers in diagnostic procedures. But up to now, there are no reliable biomarkers of major depressive disorder (MDD) and other types of depression.

Objective: The purpose of this review is to find fundamentals in pathological mechanisms of depression, which could be a basis for development of molecular and genetic biomarkers, being the most feasible for clinical use.

Method: This review summarizes the published data using PubMed, Science Direct, Google Scholar and Scopus.

Results: In this review, we summarized and discussed findings in molecular biology, genetics, neuroplasticity, neurotransmitters, and neuroimaging that could increase our understanding of the biological foundations of depression and show new directions for the development of reliable biomarkers. We did not find any molecular and genetic biomarker approved for the clinic. But the Genome-Wide Association Study method promises some progress in the development of biomarkers based on SNP in the future. Epigenetic factors also are a promising target for biomarkers. We have found some differences in the etiology of different types of atypical and melancholic depression. This knowledge could be the basis for development of biomarkers for clinical practice in diagnosis, prognosis and selection of treatment.

Conclusion: Depression is not a monoetiological disease. Many pathological mechanisms are involved in depression, thus up to now, there is no approved and reliable biomarker for diagnosis, prognosis and correction of treatment of depression. The structural and functional complexity of the brain, the lack of invasive technology, poor correlations between genetic and clinical manifestation of depression, imperfect psychiatric classification and taxonomy of subtypes of disease are the main causes of this situation. One of the possible ways to come over this situation can be to pay attention to the trigger mechanism of disease and its subtypes. Researchers and clinicians should focus their efforts on searching the trigger mechanism of depression and different types of it . HPA axis can be a candidate for such trigger in depression caused by stress, because it influences the main branches of disease: neuroinflammation, activity of biogenic amines, oxidative and nitrosative stress, epigenetic factors, metabolomics, etc. But before we shall find any trigger mechanism, we need to create complex biomarkers reflecting genetic, epigenetic, metabolomics and other pathological changes in different types of depression. Recently the most encouraging results have been obtained from genetics and neuroimaging. Continuing research in these areas should be forced by using computational, statistical and systems biology approaches, which can allow to obtain more knowledge about the neurobiology of depression. In order to obtain clinically useful tests, search for biomarkers should use appropriate research methodologies with increasing samples and identifying more homogeneous groups of depressed patients.

Keywords: Depression, biomarkers, stress, neuroinflammation, neuroplasticity, neuroimaging, genetics, pharmacology.

[1]
Vos T, Abajobir A, Abbafati C, et al. Prev GDII. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet 2017; 390: 1211-59.
[http://dx.doi.org/10.1016/S0140-6736(17)32154-2]
[2]
Greenberg PE, Fournier AA, Sisitsky T, Pike CT, Kessler RC. The economic burden of adults with major depressive disorder in the United States (2005 and 2010). J Clin Psychiatry 2015; 76(2): 155-62.
[http://dx.doi.org/10.4088/JCP.14m09298] [PMID: 25742202]
[3]
Cacabelos R, Torrellas C, Fernández-Novoa L, Aliev G. Neuroimmune Crosstalk in CNS Disorders: The Histamine Connection. Curr Pharm Des 2016; 22(7): 819-48.
[http://dx.doi.org/10.2174/1381612822666151209150954] [PMID: 26648474]
[4]
Drysdale AT, Grosenick L, Downar J, et al. Resting-state connectivity biomarkers define neurophysiological subtypes of depression. Nat Med 2017; 23(1): 28-38.
[http://dx.doi.org/10.1038/nm.4246] [PMID: 27918562]
[5]
den Braber A, Bohlken MM, Brouwer RM, et al. Heritability of subcortical brain measures: a perspective for future genome-wide association studies. Neuroimage 2013; 83: 98-102.
[http://dx.doi.org/10.1016/j.neuroimage.2013.06.027] [PMID: 23770413]
[6]
Frances A. The new crisis of confidence in psychiatric diagnosis. Ann Intern Med 2013; 159(3): 221-2.
[PMID: 23685989]
[7]
Redei EE, Andrus BM, Kwasny MJ, et al. Blood transcriptomic biomarkers in adult primary care patients with major depressive disorder undergoing cognitive behavioral therapy. Transl Psychiatry 2014; 4e442
[http://dx.doi.org/10.1038/tp.2014.66] [PMID: 25226551]
[8]
Alawieh A, Zaraket FA, Li JL, et al. Systems biology, bioinformatics, and biomarkers in neuropsychiatry. Front Neurosci 2012; 6: 187.
[http://dx.doi.org/10.3389/fnins.2012.00187] [PMID: 23269912]
[9]
Yee CM, Javitt DC, Miller GA. Replacing DSM Categorical Analyses With Dimensional Analyses in Psychiatry Research: The Research Domain Criteria Initiative. JAMA Psychiatry 2015; 72(12): 1159-60.
[http://dx.doi.org/10.1001/jamapsychiatry.2015.1900] [PMID: 26559005]
[10]
Group BDW, Atkinson AJ Jr, Colburn WA, et al. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 2001; 69(3): 89-95.
[http://dx.doi.org/10.1067/mcp.2001.113989] [PMID: 11240971]
[11]
Niculescu AB, Le-Niculescu H. Convergence of recent GWAS data for suicidality with previous blood biomarkers: independent reproducibility using independent methodologies in independent cohorts. Mol Psychiatry 2020; 25(1): 19-21.
[http://dx.doi.org/10.1038/s41380-019-0465-6] [PMID: 31383925]
[12]
Chen JJ, Bai SJ, Li WW, et al. Urinary biomarker panel for diagnosing patients with depression and anxiety disorders. Transl Psychiatry 2018; 8(1): 192.
[http://dx.doi.org/10.1038/s41398-018-0245-0] [PMID: 30232320]
[13]
Knorr U, Vinberg M, Kessing L, Wetterslev J. Salivary cortisol in depressed patients versus control persons: a systematic review and meta-analysis (vol 35, pg 1275, 2010). Psychoneuroendocrinology 2011; 36: 1427-9.
[http://dx.doi.org/10.1016/j.psyneuen.2011.08.006]
[14]
Sundberg I, Ramklint M, Stridsberg M, Papadopoulos FC, Ekselius L, Cunningham JL. Salivary Melatonin in Relation to Depressive Symptom Severity in Young Adults. PLoS One 2016; 11(4)e0152814
[http://dx.doi.org/10.1371/journal.pone.0152814] [PMID: 27042858]
[15]
Nandam LS, Brazel M, Zhou M, Jhaveri DJ. Cortisol and Major Depressive Disorder-Translating Findings From Humans to Animal Models and Back. Front Psychiatry 2020; 10: 974.
[http://dx.doi.org/10.3389/fpsyt.2019.00974] [PMID: 32038323]
[16]
Carroll BJ, Curtis GC, Mendels J. Neuroendocrine regulation in depression. II. Discrimination of depressed from nondepressed patients. Arch Gen Psychiatry 1976; 33(9): 1051-8.
[http://dx.doi.org/10.1001/archpsyc.1976.01770090041003] [PMID: 962489]
[17]
Zobel AW, Nickel T, Sonntag A, Uhr M, Holsboer F, Ising M. Cortisol response in the combined dexamethasone/CRH test as predictor of relapse in patients with remitted depression. a prospective study. J Psychiatr Res 2001; 35(2): 83-94.
[http://dx.doi.org/10.1016/S0022-3956(01)00013-9] [PMID: 11377437]
[18]
Watson S, Gallagher P, Del-Estal D, Hearn A, Ferrier IN, Young AH. Hypothalamic-pituitary-adrenal axis function in patients with chronic depression. Psychol Med 2002; 32(6): 1021-8.
[http://dx.doi.org/10.1017/S0033291702005998] [PMID: 12214783]
[19]
Lamers F, Vogelzangs N, Merikangas KR, de Jonge P, Beekman AT, Penninx BW. Evidence for a differential role of HPA-axis function, inflammation and metabolic syndrome in melancholic versus atypical depression. Mol Psychiatry 2013; 18(6): 692-9.
[http://dx.doi.org/10.1038/mp.2012.144] [PMID: 23089630]
[20]
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(2): 321-38.
[http://dx.doi.org/10.1038/s41380-019-0585-z] [PMID: 31745238]
[21]
Burke HM, Davis MC, Otte C, Mohr DC. Depression and cortisol responses to psychological stress: a meta-analysis. Psychoneuroendocrinology 2005; 30(9): 846-56.
[http://dx.doi.org/10.1016/j.psyneuen.2005.02.010] [PMID: 15961250]
[22]
Menke A, Arloth J, Best J, et al. Time-dependent effects of dexamethasone plasma concentrations on glucocorticoid receptor challenge tests. Psychoneuroendocrinology 2016; 69: 161-71.
[http://dx.doi.org/10.1016/j.psyneuen.2016.04.003] [PMID: 27107207]
[23]
Hellhammer DH, Wüst S, Kudielka BM. Salivary cortisol as a biomarker in stress research. Psychoneuroendocrinology 2009; 34(2): 163-71.
[http://dx.doi.org/10.1016/j.psyneuen.2008.10.026] [PMID: 19095358]
[24]
Bao AM, Meynen G, Swaab DF. The stress system in depression and neurodegeneration: focus on the human hypothalamus. Brain Res Brain Res Rev 2008; 57(2): 531-53.
[http://dx.doi.org/10.1016/j.brainresrev.2007.04.005] [PMID: 17524488]
[25]
Dinan TG, Scott LV. Anatomy of melancholia: focus on hypothalamic-pituitary-adrenal axis overactivity and the role of vasopressin. J Anat 2005; 207(3): 259-64.
[http://dx.doi.org/10.1111/j.1469-7580.2005.00443.x] [PMID: 16185250]
[26]
Peixoto C, Grande AJ, Mallmann MB, Nardi AE, Cardoso A, Veras AB. Dehydroepiandrosterone (DHEA) for Depression: A Systematic Review and Meta-Analysis. CNS Neurol Disord Drug Targets 2018; 17(9): 706-11.
[http://dx.doi.org/10.2174/1871527317666180817153914] [PMID: 30124161]
[27]
Wolkowitz OM, Reus VI, Keebler A, et al. Double-blind treatment of major depression with dehydroepiandrosterone. Am J Psychiatry 1999; 156(4): 646-9.
[PMID: 10200751]
[28]
Binder G, Weber S, Ehrismann M, et al. South German Working Group for Pediatric Endocrinology. Effects of dehydroepiandrosterone therapy on pubic hair growth and psychological well-being in adolescent girls and young women with central adrenal insufficiency: a double-blind, randomized, placebo-controlled phase III trial. J Clin Endocrinol Metab 2009; 94(4): 1182-90.
[http://dx.doi.org/10.1210/jc.2008-1982] [PMID: 19126625]
[29]
Mocking RJ, Pellikaan CM, Lok A, et al. DHEAS and cortisol/DHEAS-ratio in recurrent depression: State, or trait predicting 10-year recurrence? Psychoneuroendocrinology 2015; 59: 91-101.
[http://dx.doi.org/10.1016/j.psyneuen.2015.05.006] [PMID: 26036454]
[30]
Strous RD, Maayan R, Kotler M, Weizman A. Hormonal profile effects following dehydroepiandrosterone (DHEA) administration to schizophrenic patients. Clin Neuropharmacol 2005; 28(6): 265-9.
[http://dx.doi.org/10.1097/01.wnf.0000188716.25211.58] [PMID: 16340380]
[31]
Maes M, Bosmans E, De Jongh R, Kenis G, Vandoolaeghe E, Neels H. Increased serum IL-6 and IL-1 receptor antagonist concentrations in major depression and treatment resistant depression. Cytokine 1997; 9(11): 853-8.
[http://dx.doi.org/10.1006/cyto.1997.0238] [PMID: 9367546]
[32]
Rosenblat JD, Cha DS, Mansur RB, McIntyre RS. Inflamed moods: a review of the interactions between inflammation and mood disorders. Prog Neuropsychopharmacol Biol Psychiatry 2014; 53: 23-34.
[http://dx.doi.org/10.1016/j.pnpbp.2014.01.013] [PMID: 24468642]
[33]
Raison CL, Capuron L, Miller AH. Cytokines sing the blues: inflammation and the pathogenesis of depression. Trends Immunol 2006; 27(1): 24-31.
[http://dx.doi.org/10.1016/j.it.2005.11.006] [PMID: 16316783]
[34]
Hodes GE, Kana V, Menard C, Merad M, Russo SJ. Neuroimmune mechanisms of depression. Nat Neurosci 2015; 18(10): 1386-93.
[http://dx.doi.org/10.1038/nn.4113] [PMID: 26404713]
[35]
Glaus J, von Känel R, Lasserre AM, et al. Mood disorders and circulating levels of inflammatory markers in a longitudinal population-based study. Psychol Med 2018; 48(6): 961-73.
[http://dx.doi.org/10.1017/S0033291717002744] [PMID: 28929992]
[36]
Rudaz DA, Vandeleur CL, Gebreab SZ, et al. Partially distinct combinations of psychological, metabolic and inflammatory risk factors are prospectively associated with the onset of the subtypes of Major Depressive Disorder in midlife. J Affect Disord 2017; 222: 195-203.
[http://dx.doi.org/10.1016/j.jad.2017.07.016] [PMID: 28710953]
[37]
Wiener CD, Moreira FP, Portela LV, et al. Interleukin-6 and Interleukin-10 in mood disorders: A population-based study. Psychiatry Res 2019; 273: 685-9.
[http://dx.doi.org/10.1016/j.psychres.2019.01.100] [PMID: 31207853]
[38]
Gazal M, Jansen K, Souza LD, et al. Association of interleukin-10 levels with age of onset and duration of illness in patients with major depressive disorder. Br J Psychiatry 2015; 37(4): 296-302.
[http://dx.doi.org/10.1590/1516-4446-2014-1452] [PMID: 26421934]
[39]
Yamasaki K, Hasegawa T, Takeda M. Serum level of soluble interleukin 6 receptor is a useful biomarker for identification of treatment-resistant major depressive disorder. Neuropsychopharmacology Reports 2020; 40(2): 130-7.
[http://dx.doi.org/10.1002/npr2.12100]
[40]
Köhler CA, Freitas TH, Maes M, et al. Peripheral cytokine and chemokine alterations in depression: a meta-analysis of 82 studies. Acta Psychiatr Scand 2017; 135(5): 373-87.
[http://dx.doi.org/10.1111/acps.12698] [PMID: 28122130]
[41]
Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol 2016; 16(1): 22-34.
[http://dx.doi.org/10.1038/nri.2015.5] [PMID: 26711676]
[42]
Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol Psychiatry 2009; 65(9): 732-41.
[http://dx.doi.org/10.1016/j.biopsych.2008.11.029] [PMID: 19150053]
[43]
Leonard BE. Inflammation and depression: a causal or coincidental link to the pathophysiology? Acta Neuropsychiatr 2018; 30(1): 1-16.
[http://dx.doi.org/10.1017/neu.2016.69] [PMID: 28112061]
[44]
Campelo SR, da Silva MB, Vieira JL, da Silva JP, Salgado CG. Effects of immunomodulatory drugs on TNF-α and IL-12 production by purified epidermal langerhans cells and peritoneal macrophages. BMC Res Notes 2011; 4: 24.
[http://dx.doi.org/10.1186/1756-0500-4-24] [PMID: 21276247]
[45]
Obuchowicz E, Kowalski J, Labuzek K, Krysiak R, Pendzich J, Herman Z. Amitriptyline and nortriptyline inhibit interleukin-1 beta and tumour necrosis factor-alpha release by rat mixed glial and microglial cell cultures. Int J Neuropsychopharmacol 2006; 9: 27-35.
[http://dx.doi.org/10.1017/S146114570500547X] [PMID: 15963243]
[46]
Latendresse G, Ruiz RJ, Wong B. Psychological distress and SSRI use predict variation in inflammatory cytokines during pregnancy. Open J Obstet Gynecol 2013; 3(1A): 184-91.
[http://dx.doi.org/10.4236/ojog.2013.31A034] [PMID: 24524011]
[47]
Hannestad J, DellaGioia N, Bloch M. The effect of antidepressant medication treatment on serum levels of inflammatory cytokines: a meta-analysis. Neuropsychopharmacology 2011; 36(12): 2452-9.
[http://dx.doi.org/10.1038/npp.2011.132] [PMID: 21796103]
[48]
Vogelzangs N, Duivis HE, Beekman ATF, et al. Association of depressive disorders, depression characteristics and antidepressant medication with inflammation. Transl Psychiatry 2012; 2: e79-9.
[http://dx.doi.org/10.1038/tp.2012.8] [PMID: 22832816]
[49]
Chocano-Bedoya PO, Mirzaei F, O’Reilly EJ, et al. C-reactive protein, interleukin-6, soluble tumor necrosis factor α receptor 2 and incident clinical depression. J Affect Disord 2014; 163: 25-32.
[http://dx.doi.org/10.1016/j.jad.2014.03.023] [PMID: 24836084]
[50]
Copeland WE, Shanahan L, Worthman C, Angold A, Costello EJ. Cumulative depression episodes predict later C-reactive protein levels: a prospective analysis. Biol Psychiatry 2012; 71(1): 15-21.
[http://dx.doi.org/10.1016/j.biopsych.2011.09.023] [PMID: 22047718]
[51]
Carvalho AF, Köhler CA, McIntyre RS, et al. Peripheral vascular endothelial growth factor as a novel depression biomarker: A meta-analysis. Psychoneuroendocrinology 2015; 62: 18-26.
[http://dx.doi.org/10.1016/j.psyneuen.2015.07.002] [PMID: 26210676]
[52]
Harley J, Luty S, Carter J, Mulder R, Joyce P. Elevated C-reactive protein in depression: a predictor of good long-term outcome with antidepressants and poor outcome with psychotherapy. J Psychopharmacol 2010; 24(4): 625-6.
[http://dx.doi.org/10.1177/0269881109102770] [PMID: 19282426]
[53]
Chang HH, Lee IH, Gean PW, et al. Treatment response and cognitive impairment in major depression: association with C-reactive protein. Brain Behav Immun 2012; 26(1): 90-5.
[http://dx.doi.org/10.1016/j.bbi.2011.07.239] [PMID: 21839826]
[54]
Petralia MC, Mazzon E, Fagone P, et al. Pathogenic contribution of the Macrophage migration inhibitory factor family to major depressive disorder and emerging tailored therapeutic approaches. J Affect Disord 2020; 263: 15-24.
[http://dx.doi.org/10.1016/j.jad.2019.11.127] [PMID: 31818772]
[55]
Wang X, Sundquist K, Palmér K, Hedelius A, Memon AA, Sundquist J. Macrophage Migration Inhibitory Factor and microRNA-451a in Response to Mindfulness-based Therapy or Treatment as Usual in Patients with Depression, Anxiety, or Stress and Adjustment Disorders. Int J Neuropsychopharmacol 2018; 21(6): 513-21.
[http://dx.doi.org/10.1093/ijnp/pyy001] [PMID: 29373661]
[56]
Lin P, Ding B, Wu Y, Dong K, Li Q. Mitogen-stimulated cell proliferation and cytokine production in major depressive disorder patients. BMC Psychiatry 2018; 18(1): 330.
[http://dx.doi.org/10.1186/s12888-018-1906-5] [PMID: 30314474]
[57]
Hall JR, Wiechmann A, Edwards M, Johnson LA, O’Bryant SE. IL-7 and Depression: The importance of gender and blood fraction. Behav Brain Res 2016; 315: 147-9.
[http://dx.doi.org/10.1016/j.bbr.2016.08.026] [PMID: 27555535]
[58]
Stelzhammer V, Haenisch F, Chan MK, et al. Proteomic changes in serum of first onset, antidepressant drug-naïve major depression patients. Int J Neuropsychopharmacol 2014; 17(10): 1599-608.
[http://dx.doi.org/10.1017/S1461145714000819] [PMID: 24901538]
[59]
Shinko Y, Otsuka I, Okazaki S, et al. Chemokine alterations in the postmortem brains of suicide completers. J Psychiatr Res 2020; 120: 29-33.
[http://dx.doi.org/10.1016/j.jpsychires.2019.10.008] [PMID: 31629996]
[60]
Toft H, Neupane SP, Bramness JG, Tilden T, Wampold BE, Lien L. The effect of trauma and alcohol on the relationship between level of cytokines and depression among patients entering psychiatric treatment. BMC Psychiatry 2018; 18(1): 95.
[http://dx.doi.org/10.1186/s12888-018-1677-z] [PMID: 29631540]
[61]
Milenkovic VM, Sarubin N, Hilbert S, et al. Macrophage-Derived Chemokine: A Putative Marker of Pharmacological Therapy Response in Major Depression? Neuroimmunomodulation 2017; 24(2): 106-12.
[http://dx.doi.org/10.1159/000479739] [PMID: 28898872]
[62]
Mindt S, Neumaier M, Hoyer C, Sartorius A, Kranaster L. Cytokine-mediated cellular immune activation in electroconvulsive therapy: A CSF study in patients with treatment-resistant depression. World J Biol Psychiatry 2020; 21(2): 139-47.
[http://dx.doi.org/10.1080/15622975.2019.1618494] [PMID: 31081432]
[63]
Ozpercin PU, Kendirlioglu BK, Sozen S, et al. Decreased circulating urokinase plasminogen activator receptor (uPAR) concentration in acute episodes of bipolar disorder; could it be a reflection of axonal injury? Psychoneuroendocrinology 2018; 90: 122-6.
[http://dx.doi.org/10.1016/j.psyneuen.2018.02.009] [PMID: 29482134]
[64]
Gustafsson A, Ventorp F, Wisén AG, Ohlsson L, Ljunggren L, Westrin Å. Effects of Acute Exercise on Circulating Soluble Form of the Urokinase Receptor in Patients With Major Depressive Disorder. Biomark Insights 2017; 121177271917704193
[http://dx.doi.org/10.1177/1177271917704193] [PMID: 28469403]
[65]
Ventorp F, Gustafsson A, Träskman-Bendz L, Westrin Å, Ljunggren L. Increased Soluble Urokinase-Type Plasminogen Activator Receptor (suPAR) Levels in Plasma of Suicide Attempters. PLoS One 2015; 10(10)e0140052
[http://dx.doi.org/10.1371/journal.pone.0140052] [PMID: 26451727]
[66]
Bocchio-Chiavetto L, Bagnardi V, Zanardini R, et al. Serum and plasma BDNF levels in major depression: a replication study and meta-analyses. World J Biol Psychiatry 2010; 11(6): 763-73.
[http://dx.doi.org/10.3109/15622971003611319] [PMID: 20334574]
[67]
Molendijk ML, Spinhoven P, Polak M, Bus BA, Penninx BW, Elzinga BM. Serum BDNF concentrations as peripheral manifestations of depression: evidence from a systematic review and meta-analyses on 179 associations (N=9484). Mol Psychiatry 2014; 19(7): 791-800.
[http://dx.doi.org/10.1038/mp.2013.105] [PMID: 23958957]
[68]
Nase S, Köhler S, Jennebach J, et al. Role of serum brain derived neurotrophic factor and central N-acetylaspartate for clinical response under antidepressive pharmacotherapy. Neurosignals 2016; 24(1): 1-14.
[http://dx.doi.org/10.1159/000442607] [PMID: 26859851]
[69]
Sagud M, Nikolac Perkovic M, Vuksan-Cusa B, et al. A prospective, longitudinal study of platelet serotonin and plasma brain-derived neurotrophic factor concentrations in major depression: effects of vortioxetine treatment. Psychopharmacology (Berl) 2016; 233(17): 3259-67.
[http://dx.doi.org/10.1007/s00213-016-4364-0] [PMID: 27356518]
[70]
Youssef MM, Underwood MD, Huang Y-Y, et al. Association of BDNF Val66Met polymorphism and brain BDNF levels with major depression and suicide. Int J Neuropsychopharmacol 2018; 21(6): 528-38.
[http://dx.doi.org/10.1093/ijnp/pyy008] [PMID: 29432620]
[71]
Molendijk ML, Bus BA, Spinhoven P, et al. Serum levels of brain-derived neurotrophic factor in major depressive disorder: state-trait issues, clinical features and pharmacological treatment. Mol Psychiatry 2011; 16(11): 1088-95.
[http://dx.doi.org/10.1038/mp.2010.98] [PMID: 20856249]
[72]
Polacchini A, Metelli G, Francavilla R, et al. A method for reproducible measurements of serum BDNF: comparison of the performance of six commercial assays. Sci Rep 2015; 5: 17989.
[http://dx.doi.org/10.1038/srep17989] [PMID: 26656852]
[73]
Airaksinen MS, Saarma M. The GDNF family: signalling, biological functions and therapeutic value. Nat Rev Neurosci 2002; 3(5): 383-94.
[http://dx.doi.org/10.1038/nrn812] [PMID: 11988777]
[74]
Liu Q, Zhu HY, Li B, Wang YQ, Yu J, Wu GC. Chronic clomipramine treatment restores hippocampal expression of glial cell line-derived neurotrophic factor in a rat model of depression. J Affect Disord 2012; 141(2-3): 367-72.
[http://dx.doi.org/10.1016/j.jad.2012.03.018] [PMID: 22658339]
[75]
Anisman H, Du L, Palkovits M, et al. Serotonin receptor subtype and p11 mRNA expression in stress-relevant brain regions of suicide and control subjects. J Psychiatry Neurosci 2008; 33(2): 131-41.
[PMID: 18330459]
[76]
Svenningsson P, Kim Y, Warner-Schmidt J, Oh YS, Greengard P. p11 and its role in depression and therapeutic responses to antidepressants. Nat Rev Neurosci 2013; 14(10): 673-80.
[http://dx.doi.org/10.1038/nrn3564] [PMID: 24002251]
[77]
Jin J, Bhatti DL, Lee K-W, et al. Ahnak scaffolds p11/Anxa2 complex and L-type voltage-gated calcium channel and modulates depressive behavior. Mol Psychiatry 2020; 25(5): 1035-49.
[http://dx.doi.org/10.1038/s41380-019-0371-y] [PMID: 30760886]
[78]
Warner-Schmidt JL, Duman RS. VEGF as a potential target for therapeutic intervention in depression. Curr Opin Pharmacol 2008; 8(1): 14-9.
[http://dx.doi.org/10.1016/j.coph.2007.10.013] [PMID: 18061540]
[79]
Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science 2003; 301(5631): 386-9.
[http://dx.doi.org/10.1126/science.1083968] [PMID: 12869766]
[80]
Sharma AN. da Costa e Silva BF, Soares JC, Carvalho AF, Quevedo J. Role of trophic factors GDNF, IGF-1 and VEGF in major depressive disorder: A comprehensive review of human studies. J Affect Disord 2016; 197: 9-20.
[http://dx.doi.org/10.1016/j.jad.2016.02.067] [PMID: 26956384]
[81]
Deng Z, Deng S, Zhang MR, Tang MM. Fibroblast Growth Factors in Depression. Front Pharmacol 2019; 10: 60.
[http://dx.doi.org/10.3389/fphar.2019.00060] [PMID: 30804785]
[82]
Fonseka TM, MacQueen GM, Kennedy SH. Neuroimaging biomarkers as predictors of treatment outcome in Major Depressive Disorder. J Affect Disord 2018; 233: 21-35.
[http://dx.doi.org/10.1016/j.jad.2017.10.049] [PMID: 29150145]
[83]
McGrath CL, Kelley ME, Holtzheimer PE, et al. Toward a neuroimaging treatment selection biomarker for major depressive disorder. JAMA Psychiatry 2013; 70(8): 821-9.
[http://dx.doi.org/10.1001/jamapsychiatry.2013.143] [PMID: 23760393]
[84]
Hasler G, van der Veen JW, Tumonis T, Meyers N, Shen J, Drevets WC. Reduced prefrontal glutamate/glutamine and γ-aminobutyric acid levels in major depression determined using proton magnetic resonance spectroscopy. Arch Gen Psychiatry 2007; 64(2): 193-200.
[http://dx.doi.org/10.1001/archpsyc.64.2.193] [PMID: 17283286]
[85]
Urrila AS, Hakkarainen A, Castaneda A, Paunio T, Marttunen M, Lundbom N. Frontal cortex myo-inositol is associated with sleep and depression in adolescents: A proton magnetic resonance spectroscopy study. Neuropsychobiology 2017; 75(1): 21-31.
[http://dx.doi.org/10.1159/000478861] [PMID: 28793304]
[86]
Godlewska BR, Near J, Cowen PJ. Neurochemistry of major depression: a study using magnetic resonance spectroscopy. Psychopharmacology (Berl) 2015; 232(3): 501-7.
[http://dx.doi.org/10.1007/s00213-014-3687-y] [PMID: 25074444]
[87]
Morris G, Anderson G, Dean O, et al. The glutathione system: a new drug target in neuroimmune disorders. Mol Neurobiol 2014; 50(3): 1059-84.
[http://dx.doi.org/10.1007/s12035-014-8705-x] [PMID: 24752591]
[88]
Sanacora G, Treccani G, Popoli M. Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology 2012; 62(1): 63-77.
[http://dx.doi.org/10.1016/j.neuropharm.2011.07.036] [PMID: 21827775]
[89]
McOmish CE, Pavey G, Gibbons A, et al. Lower [3H]LY341495 binding to mGlu2/3 receptors in the anterior cingulate of subjects with major depressive disorder but not bipolar disorder or schizophrenia. J Affect Disord 2016; 190: 241-8.
[http://dx.doi.org/10.1016/j.jad.2015.10.004] [PMID: 26521087]
[90]
Matosin N, Fernandez-Enright F, Frank E, et al. Metabotropic glutamate receptor mGluR2/3 and mGluR5 binding in the anterior cingulate cortex in psychotic and nonpsychotic depression, bipolar disorder and schizophrenia: implications for novel mGluR-based therapeutics. J Psychiatry Neurosci 2014; 39(6): 407-16.
[http://dx.doi.org/10.1503/jpn.130242] [PMID: 24949866]
[91]
Luykx JJ, Laban KG, van den Heuvel MP, et al. Region and state specific glutamate downregulation in major depressive disorder: a meta-analysis of (1)H-MRS findings. Neurosci Biobehav Rev 2012; 36(1): 198-205.
[http://dx.doi.org/10.1016/j.neubiorev.2011.05.014] [PMID: 21672551]
[92]
Adell A. Brain NMDA Receptors in Schizophrenia and Depression. Biomolecules 2020; 10(6): 10.
[http://dx.doi.org/10.3390/biom10060947] [PMID: 32585886]
[93]
Lee PH, Perlis RH, Jung JY, et al. Multi-locus genome-wide association analysis supports the role of glutamatergic synaptic transmission in the etiology of major depressive disorder. Transl Psychiatry 2012; 2e184
[http://dx.doi.org/10.1038/tp.2012.95] [PMID: 23149448]
[94]
Lin CH, Huang MW, Lin CH, Huang CH, Lane HY. Altered mRNA expressions for N-methyl-D-aspartate receptor-related genes in WBC of patients with major depressive disorder. J Affect Disord 2019; 245: 1119-25.
[http://dx.doi.org/10.1016/j.jad.2018.12.016] [PMID: 30699855]
[95]
Cole J, Costafreda SG, McGuffin P, Fu CH. Hippocampal atrophy in first episode depression: a meta-analysis of magnetic resonance imaging studies. J Affect Disord 2011; 134(1-3): 483-7.
[http://dx.doi.org/10.1016/j.jad.2011.05.057] [PMID: 21745692]
[96]
Zhuo C, Li G, Lin X, et al. The rise and fall of MRI studies in major depressive disorder. Transl Psychiatry 2019; 9(1): 335.
[http://dx.doi.org/10.1038/s41398-019-0680-6] [PMID: 31819044]
[97]
Castanheira L, Silva C, Cheniaux E, Telles-Correia D. Neuroimaging correlates of depression-implications to clinical practice. Front Psychiatry 2019; 10: 703.
[http://dx.doi.org/10.3389/fpsyt.2019.00703] [PMID: 31632306]
[98]
Han MR, Han KM, Kim A, et al. Whole-exome sequencing identifies variants associated with structural MRI markers in patients with bipolar disorders. J Affect Disord 2019; 249: 159-68.
[http://dx.doi.org/10.1016/j.jad.2019.02.028] [PMID: 30772743]
[99]
Kandilarova S, Stoyanov D, Sirakov N, Maes M, Specht K. Reduced grey matter volume in frontal and temporal areas in depression: contributions from voxel-based morphometry study. Acta Neuropsychiatr 2019; 31(5): 252-7.
[http://dx.doi.org/10.1017/neu.2019.20] [PMID: 31234950]
[100]
Arnone D. Functional MRI findings, pharmacological treatment in major depression and clinical response. Prog Neuropsychopharmacol Biol Psychiatry 2019; 91: 28-37.
[http://dx.doi.org/10.1016/j.pnpbp.2018.08.004] [PMID: 30099082]
[101]
Kandilarova S, Stoyanov D, Kostianev S, Specht K. Altered Resting State Effective Connectivity of Anterior Insula in Depression. Front Psychiatry 2018; 9: 83.
[http://dx.doi.org/10.3389/fpsyt.2018.00083] [PMID: 29599728]
[102]
Dubol M, Trichard C, Leroy C, et al. Lower midbrain dopamine transporter availability in depressed patients: Report from high-resolution PET imaging. J Affect Disord 2020; 262: 273-7.
[http://dx.doi.org/10.1016/j.jad.2019.10.041] [PMID: 31732277]
[103]
Rosenbaum D, Haipt A, Fuhr K, et al. Aberrant functional connectivity in depression as an index of state and trait rumination. Sci Rep 2017; 7(1): 2174.
[http://dx.doi.org/10.1038/s41598-017-02277-z] [PMID: 28526867]
[104]
Beijers L, Wardenaar KJ, van Loo HM, Schoevers RA. Data-driven biological subtypes of depression: systematic review of biological approaches to depression subtyping. Mol Psychiatry 2019; 24(6): 888-900.
[http://dx.doi.org/10.1038/s41380-019-0385-5] [PMID: 30824865]
[105]
Wray NR, Ripke S, Mattheisen M, et al. eQTLGen; 23andMe; Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat Genet 2018; 50(5): 668-81.
[http://dx.doi.org/10.1038/s41588-018-0090-3] [PMID: 29700475]
[106]
Katsuki A, Kakeda S, Watanabe K, et al. A single-nucleotide polymorphism influences brain morphology in drug-naïve patients with major depressive disorder. Neuropsychiatr Dis Treat 2019; 15: 2425-32.
[http://dx.doi.org/10.2147/NDT.S204461] [PMID: 31692503]
[107]
Igata R, Katsuki A, Kakeda S, et al. PCLO rs2522833-mediated gray matter volume reduction in patients with drug-naive, first-episode major depressive disorder. Transl Psychiatry 2017; 7(5)e1140
[http://dx.doi.org/10.1038/tp.2017.100] [PMID: 28556829]
[108]
Geschwind DH, Flint J. Genetics and genomics of psychiatric disease. Science 2015; 349(6255): 1489-94.
[http://dx.doi.org/10.1126/science.aaa8954] [PMID: 26404826]
[109]
McGuffin P, Katz R, Watkins S, Rutherford J. A hospital-based twin register of the heritability of DSM-IV unipolar depression. Arch Gen Psychiatry 1996; 53(2): 129-36.
[http://dx.doi.org/10.1001/archpsyc.1996.01830020047006] [PMID: 8629888]
[110]
Sullivan PF, Neale MC, Kendler KS. Genetic epidemiology of major depression: review and meta-analysis. Am J Psychiatry 2000; 157(10): 1552-62.
[http://dx.doi.org/10.1176/appi.ajp.157.10.1552] [PMID: 11007705]
[111]
Lee SH, Ripke S, Neale BM, et al. International Inflammatory Bowel Disease Genetics Consortium (IIBDGC). Genetic relationship between five psychiatric disorders estimated from genome-wide SNPs. Nat Genet 2013; 45(9): 984-94.
[http://dx.doi.org/10.1038/ng.2711] [PMID: 23933821]
[112]
Uher R. Gene-environment interactions in severe mental illness. Front Psychiatry 2014; 5: 48.
[http://dx.doi.org/10.3389/fpsyt.2014.00048] [PMID: 24860514]
[113]
Howard DM, Adams MJ, Clarke T-K, et al. Genome-wide meta-analysis of depression in 807,553 individuals identifies 102 independent variants with replication in a further 1,507,153 individuals. bioRxiv 2018; 12(24)433367
[114]
Ripke S, Wray NR, Lewis CM, et al. A mega-analysis of genome-wide association studies for major depressive disorder. Mol Psychiatry 2013; 18(4): 497-511.
[http://dx.doi.org/10.1038/mp.2012.21] [PMID: 22472876]
[115]
Chatterjee N, Shi J, García-Closas M. Developing and evaluating polygenic risk prediction models for stratified disease prevention. Nat Rev Genet 2016; 17(7): 392-406.
[http://dx.doi.org/10.1038/nrg.2016.27] [PMID: 27140283]
[116]
Van der Auwera S, Janowitz D, Schulz A, et al. Interaction among childhood trauma and functional polymorphisms in the serotonin pathway moderate the risk of depressive disorders. Eur Arch Psychiatry Clin Neurosci 2014; 264(Suppl. 1): S45-54.
[http://dx.doi.org/10.1007/s00406-014-0536-2] [PMID: 25214390]
[117]
Mandelli L, Serretti A. Gene environment interaction studies in depression and suicidal behavior: An update. Neurosci Biobehav Rev 2013; 37(10 Pt. 1): 2375-97.
[http://dx.doi.org/10.1016/j.neubiorev.2013.07.011] [PMID: 23886513]
[118]
Appel K, Schwahn C, Mahler J, et al. Moderation of adult depression by a polymorphism in the FKBP5 gene and childhood physical abuse in the general population. Neuropsychopharmacology 2011; 36(10): 1982-91.
[http://dx.doi.org/10.1038/npp.2011.81] [PMID: 21654733]
[119]
Van der Auwera S, Peyrot WJ, Milaneschi Y, et al. Genome-wide gene-environment interaction in depression: A systematic evaluation of candidate genes: The childhood trauma working-group of PGC-MDD. Am J Med Genet B Neuropsychiatr Genet 2018; 177(1): 40-9.
[http://dx.doi.org/10.1002/ajmg.b.32593] [PMID: 29159863]
[120]
Culverhouse RC, Saccone NL, Horton AC, et al. Collaborative meta-analysis finds no evidence of a strong interaction between stress and 5-HTTLPR genotype contributing to the development of depression. Mol Psychiatry 2018; 23(1): 133-42.
[http://dx.doi.org/10.1038/mp.2017.44] [PMID: 28373689]
[121]
Border R, Johnson EC, Evans LM, et al. No Support for Historical Candidate Gene or Candidate Gene-by-Interaction Hypotheses for Major Depression Across Multiple Large Samples. Am J Psychiatry 2019; 176(5): 376-87.
[http://dx.doi.org/10.1176/appi.ajp.2018.18070881] [PMID: 30845820]
[122]
Sullivan PF, Geschwind DH. Defining the Genetic, Genomic, Cellular, and Diagnostic Architectures of Psychiatric Disorders. Cell 2019; 177(1): 162-83.
[http://dx.doi.org/10.1016/j.cell.2019.01.015] [PMID: 30901538]
[123]
Fan T, Hu Y, Xin J, Zhao M, Wang J. Analyzing the genes and pathways related to major depressive disorder via a systems biology approach. Brain Behav 2020; 10(2)e01502
[http://dx.doi.org/10.1002/brb3.1502] [PMID: 31875662]
[124]
Hyde CL, Nagle MW, Tian C, et al. Identification of 15 genetic loci associated with risk of major depression in individuals of European descent. Nat Genet 2016; 48(9): 1031-6.
[http://dx.doi.org/10.1038/ng.3623] [PMID: 27479909]
[125]
Morozov DI, Tikhonova IuG, Kinkul’kina MA, Ivanets NN. [Depression in patients with chronic hepatitis C during the antiviral treatment: a clinical and psychopathological qualification and a nosological interpretation] Zh Nevrol Psikhiatr Im S S Korsakova 2012; 112(11): 4-12.
[PMID: 23257738]
[126]
McIntosh AM, Sullivan PF, Lewis CM. Uncovering the Genetic Architecture of Major Depression. Neuron 2019; 102(1): 91-103.
[http://dx.doi.org/10.1016/j.neuron.2019.03.022] [PMID: 30946830]
[127]
Levinson DF, Mostafavi S, Milaneschi Y, et al. Genetic studies of major depressive disorder: why are there no genome-wide association study findings and what can we do about it? Biol Psychiatry 2014; 76(7): 510-2.
[http://dx.doi.org/10.1016/j.biopsych.2014.07.029] [PMID: 25201436]
[128]
Peterson RE, Cai N, Dahl AW, et al. Molecular genetic analysis subdivided by adversity exposure suggests etiologic heterogeneity in major depression. Am J Psychiatry 2018; 175(6): 545-54.
[http://dx.doi.org/10.1176/appi.ajp.2017.17060621] [PMID: 29495898]
[129]
Milaneschi Y, Lamers F, Peyrot W. Genetic Association of Major Depression With Atypical Features and Obesity-Related Immunometabolic Dysregulations. JAMA Psychiatry 2017; 74: 1279-9.
[http://dx.doi.org/10.1001/jamapsychiatry.2017.3016]
[130]
Power RA, Tansey KE, Buttenschøn HN, et al. CARDIoGRAM Consortium, GERAD1 Consortium. Genome-wide Association for Major Depression Through Age at Onset Stratification: Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium. Biol Psychiatry 2017; 81(4): 325-35.
[http://dx.doi.org/10.1016/j.biopsych.2016.05.010] [PMID: 27519822]
[131]
Hall LS, Adams MJ, Arnau-Soler A, et al. Genome-wide meta-analyses of stratified depression in Generation Scotland and UK Biobank. Transl Psychiatry 2018; 8(1): 9.
[http://dx.doi.org/10.1038/s41398-017-0034-1] [PMID: 29317602]
[132]
Chen D, Meng L, Pei F, Zheng Y, Leng J. A review of DNA methylation in depression. J Clin Neurosci 2017; 43: 39-46.
[http://dx.doi.org/10.1016/j.jocn.2017.05.022] [PMID: 28645747]
[133]
Lin E, Tsai SJ. Epigenetics and Depression: An Update. Psychiatry Investig 2019; 16(9): 654-61.
[http://dx.doi.org/10.30773/pi.2019.07.17.2] [PMID: 31455063]
[134]
Doherty TS, Forster A, Roth TL. Global and gene-specific DNA methylation alterations in the adolescent amygdala and hippocampus in an animal model of caregiver maltreatment Behav Brain Res 2016; 298(Pt. A): 55-61.
[http://dx.doi.org/10.1016/j.bbr.2015.05.028] [PMID: 26027495]
[135]
Blaze J, Asok A, Borrelli K, et al. Intrauterine exposure to maternal stress alters Bdnf IV DNA methylation and telomere length in the brain of adult rat offspring. Int J Dev Neurosci 2017; 62: 56-62.
[http://dx.doi.org/10.1016/j.ijdevneu.2017.03.007] [PMID: 28330827]
[136]
Blaze J, Asok A, Roth TL. Long-term effects of early-life caregiving experiences on brain-derived neurotrophic factor histone acetylation in the adult rat mPFC. Stress 2015; 18(6): 607-15.
[http://dx.doi.org/10.3109/10253890.2015.1071790] [PMID: 26305287]
[137]
Holmes L Jr, Shutman E, Chinaka C, Deepika K, Pelaez L, Dabney KW. Aberrant Epigenomic Modulation of Glucocorticoid Receptor Gene (NR3C1) in Early Life Stress and Major Depressive Disorder Correlation: Systematic Review and Quantitative Evidence Synthesis. Int J Environ Res Public Health 2019; 16(21): 16.
[http://dx.doi.org/10.3390/ijerph16214280] [PMID: 31689998]
[138]
Bakusic J, Schaufeli W, Claes S, Godderis L. Stress, burnout and depression: A systematic review on DNA methylation mechanisms. J Psychosom Res 2017; 92: 34-44.
[http://dx.doi.org/10.1016/j.jpsychores.2016.11.005] [PMID: 27998510]
[139]
Olsson CA, Foley DL, 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 2010; 83(2): 159-65.
[http://dx.doi.org/10.1016/j.biopsycho.2009.12.003] [PMID: 20018225]
[140]
Li M, D’Arcy C, Li X, Zhang T, Joober R, Meng X. What do DNA methylation studies tell us about depression? A systematic review. Transl Psychiatry 2019; 9(1): 68.
[http://dx.doi.org/10.1038/s41398-019-0412-y] [PMID: 30718449]
[141]
Zimmerman M, Posternak M, Friedman M, et al. Which factors influence psychiatrists’ selection of antidepressants? Am J Psychiatry 2004; 161(7): 1285-9.
[http://dx.doi.org/10.1176/appi.ajp.161.7.1285] [PMID: 15229063]
[142]
Bauer M, Pfennig A, Severus E, et al. World Federation of Societies of Biological Psychiatry. Task Force on Unipolar Depressive Disorders. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for biological treatment of unipolar depressive disorders, part 1: update 2013 on the acute and continuation treatment of unipolar depressive disorders. World J Biol Psychiatry 2013; 14(5): 334-85.
[http://dx.doi.org/10.3109/15622975.2013.804195] [PMID: 23879318]
[143]
Bousman CA, Jaksa P, Pantelis C. Systematic evaluation of commercial pharmacogenetic testing in psychiatry: a focus on CYP2D6 and CYP2C19 allele coverage and results reporting. Pharmacogenet Genomics 2017; 27(11): 387-93.
[http://dx.doi.org/10.1097/FPC.0000000000000303] [PMID: 28777243]
[144]
Müller DJ, Kekin I, Kao AC, Brandl EJ. Towards the implementation of CYP2D6 and CYP2C19 genotypes in clinical practice: update and report from a pharmacogenetic service clinic. Int Rev Psychiatry 2013; 25(5): 554-71.
[http://dx.doi.org/10.3109/09540261.2013.838944] [PMID: 24151801]
[145]
Fabbri C, Hosak L, Mössner R, et al. Consensus paper of the WFSBP Task Force on Genetics: Genetics, epigenetics and gene expression markers of major depressive disorder and antidepressant response. World J Biol Psychiatry 2017; 18(1): 5-28.
[http://dx.doi.org/10.1080/15622975.2016.1208843] [PMID: 27603714]
[146]
van Loo HM, de Jonge P, Romeijn JW, Kessler RC, Schoevers RA. Data-driven subtypes of major depressive disorder: a systematic review. BMC Med 2012; 10: 156.
[http://dx.doi.org/10.1186/1741-7015-10-156] [PMID: 23210727]
[147]
Rush AJ. The varied clinical presentations of major depressive disorder. J Clin Psychiatry 2007; 68(Suppl. 8): 4-10.
[PMID: 17640152]
[148]
Harald B, Gordon P. Meta-review of depressive subtyping models. J Affect Disord 2012; 139(2): 126-40.
[http://dx.doi.org/10.1016/j.jad.2011.07.015] [PMID: 21885128]
[149]
Cuthbert BN. The RDoC framework: facilitating transition from ICD/DSM to dimensional approaches that integrate neuroscience and psychopathology. World Psychiatry 2014; 13(1): 28-35.
[http://dx.doi.org/10.1002/wps.20087] [PMID: 24497240]
[150]
Maj M. Keeping an open attitude towards the RDoC project. World Psychiatry 2014; 13(1): 1-3.
[http://dx.doi.org/10.1002/wps.20111] [PMID: 24497235]
[151]
Robinaugh DJ, Hoekstra RHA, Toner ER, Borsboom D. The network approach to psychopathology: a review of the literature 2008-2018 and an agenda for future research. Psychol Med 2020; 50(3): 353-66.
[http://dx.doi.org/10.1017/S0033291719003404] [PMID: 31875792]
[152]
Ruggero CJ, Kotov R, Hopwood CJ, et al. Integrating the Hierarchical Taxonomy of Psychopathology (HiTOP) into clinical practice. J Consult Clin Psychol 2019; 87(12): 1069-84.
[http://dx.doi.org/10.1037/ccp0000452] [PMID: 31724426]
[153]
Ahmed AT, Frye MA, Rush AJ, et al. Mood Disorders Precision Medicine Consortium (MDPMC). Mapping depression rating scale phenotypes onto research domain criteria (RDoC) to inform biological research in mood disorders. J Affect Disord 2018; 238: 1-7.
[http://dx.doi.org/10.1016/j.jad.2018.05.005] [PMID: 29807322]
[154]
Schwabe I, Milaneschi Y, Gerring Z, et al. Unraveling the genetic architecture of major depressive disorder: merits and pitfalls of the approaches used in genome-wide association studies. Psychol Med 2019; 49(16): 2646-56.
[http://dx.doi.org/10.1017/S0033291719002502] [PMID: 31559935]
[155]
Howard DM, Adams MJ, Shirali M, et al. Genome-wide association study of depression phenotypes in UK Biobank identifies variants in excitatory synaptic pathways. Nat Commun 2018; 9: 1-10.
[156]
Muir J, Lopez J, Bagot RC. Wiring the depressed brain: optogenetic and chemogenetic circuit interrogation in animal models of depression. Neuropsychopharmacology 2019; 44(6): 1013-26.
[http://dx.doi.org/10.1038/s41386-018-0291-6] [PMID: 30555161]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy