Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-4rdrl Total loading time: 0 Render date: 2024-06-23T07:04:39.206Z Has data issue: false hasContentIssue false

Section 4 - Novel Approaches in Brain Imaging

Published online by Cambridge University Press:  12 January 2021

Sudhakar Selvaraj
Affiliation:
UTHealth School of Medicine, USA
Paolo Brambilla
Affiliation:
Università degli Studi di Milano
Jair C. Soares
Affiliation:
UT Harris County Psychiatric Center, USA
Get access
Type
Chapter
Information
Mood Disorders
Brain Imaging and Therapeutic Implications
, pp. 135 - 218
Publisher: Cambridge University Press
Print publication year: 2021

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.)

References

References

Barnett, JH, Smoller, JW. The genetics of bipolar disorder. Neuroscience. 2009; 164(1): 331343.CrossRefGoogle ScholarPubMed
McGuffin, P, Rijsdijk, F, Andrew, M et al. The heritability of bipolar affective disorder and the genetic relationship to unipolar depression. Arch Gen Psychiatry. 2003; 60(5): 497502.CrossRefGoogle ScholarPubMed
Jansen, R, Penninx, BWJH, Madar, V, et al. Gene expression in major depressive disorder. Mol Psychiatr. 2016; 21(3): 339347.Google Scholar
Hamet, P, Tremblay, J. Genetics and genomics of depression. Metabolism. 2005; 54 (5 Suppl 1): 1015.CrossRefGoogle ScholarPubMed
Chen, G, Henter, ID, Manji, HK. Translational research in bipolar disorder: Emerging insights from genetically based models. Mol Psychiatry. 2010; 15(9): 883895.CrossRefGoogle ScholarPubMed
Goes, FS. Genetics of bipolar disorder: Recent update and future directions. Psychiatr Clin North Am. 2016; 39(1): 139155.CrossRefGoogle ScholarPubMed
Bogdan, R, Salmeron, BJ, Carey, CE, et al. Imaging genetics and genomics in psychiatry: A critical review of progress and potential. Biological Psychiatry. 2017; 82(3): 165175.Google Scholar
Meyer-Lindenberg, A, Weinberger, DR. Intermediate phenotypes and genetic mechanisms of psychiatric disorders. Nat Rev Neurosci. 2006; 7(10): 818827.CrossRefGoogle ScholarPubMed
Fusar-Poli, P, Howes, O, Bechdolf, A, Borgwardt, S. Mapping vulnerability to bipolar disorder: a systematic review and meta-analysis of neuroimaging studies. J Psychiatry Neurosci. 2012; 37(3): 170184.Google Scholar
Piguet, C, Fodoulian, L, Aubry, JM, Vuilleumier, P, Houenou, J. Bipolar disorder: Functional neuroimaging markers in relatives. Neurosci Biobehav Rev. 2015; 57: 284296.Google Scholar
Dima, D, Roberts, RE, Frangou, S. Connectomic markers of disease expression, genetic risk and resilience in bipolar disorder. Transl Psychiatry. 2016; 6: e706.Google Scholar
Amico, F, Meisenzahl, E, Koutsouleris, N, et al. Structural MRI correlates for vulnerability and resilience to major depressive disorder. J Psychiatry Neurosci. 2011; 36(1): 1522.Google Scholar
Huang, H, Fan, X, Williamson, DE, Rao, U. White matter changes in healthy adolescents at familial risk for unipolar depression: A diffusion tensor imaging study. Neuropsychopharmacology. 2011; 36(3): 684691.Google Scholar
Kerner, B. Toward a deeper understanding of the genetics of bipolar disorder. Frontiers in Psychiatry. 2015; 6: 105.CrossRefGoogle Scholar
Sullivan, P, Wray, N, Consortium, PM. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depressive disorder. Eur Neuropsychopharm. 2019; 29: S805.Google Scholar
Direk, N, Williams, S, Smith, JA, et al. An analysis of two genome-wide association meta-analyses identifies a new locus for broad depression phenotype. Biol Psychiat. 2017; 82(5): 322329.Google Scholar
Hyde, CL, Nagle, MW, Tian, C, et al. Identification of 15 genetic loci associated with risk of major depression in individuals of European descent. Nature Genetics. 2016 September; 48(9): 10311036.Google Scholar
Ikeda, M, Saito, T, Kondo, K, Iwata, N. Genome-wide association studies of bipolar disorder: A systematic review of recent findings and their clinical implications. Psychiatry Clin Neurosci. 2018; 72(2): 5263.Google Scholar
Leal-Ortiz, S, Waites, CL, Terry-Lorenzo, R, et al. Piccolo modulation of Synapsin1a dynamics regulates synaptic vesicle exocytosis. Journal of Cell Biology. 2008; 181(5): 831846.CrossRefGoogle ScholarPubMed
Schuhmacher, A, Mossner, R, Hofels, S, et al. PCLO rs2522833 modulates HPA system response to antidepressant treatment in major depressive disorder. Int J Neuropsychoph. 2011; 14(2): 237245.CrossRefGoogle ScholarPubMed
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 May 30; 7(5): e1140.Google Scholar
Ryan, J, Artero, S, Carriere, I, et al. GWAS-identified risk variants for major depressive disorder: Preliminary support for an association with late-life depressive symptoms and brain structural alterations. Eur Neuropsychopharm. 2016; 26(1): 113125.CrossRefGoogle ScholarPubMed
Woudstra, S, Bochdanovits, Z, van Tol, MJ, et al. Piccolo genotype modulates neural correlates of emotion processing but not executive functioning. Transl Psychiatry. 2012; 2: e99.CrossRefGoogle Scholar
Woudstra, S, van Tol, MJ, Bochdanovits, Z, et al. Modulatory effects of the piccolo genotype on emotional memory in health and depression. PLoS One. 2013; 8(4): e61494.CrossRefGoogle ScholarPubMed
Schott, BH, Assmann, A, Schmierer, P, et al. Epistatic interaction of genetic depression risk variants in the human subgenual cingulate cortex during memory encoding. Transl Psychiatry. 2014; 4: e372.Google Scholar
Lewis, CM, Ng, MY, Butler, AW, et al. Genome-wide association study of major recurrent depression in the UK population. Am J Psychiat. 2010; 167(8): 949957.CrossRefGoogle Scholar
Bermingham, R, Carballedo, A, Lisiecka, D, et al. Effect of genetic variant in BICC1 on functional and structural brain changes in depression. Neuropsychopharmacology. 2012; 37(13): 28552862.CrossRefGoogle ScholarPubMed
Dannlowski, U, Kugel, H, Grotegerd, D, et al. NCAN cross-disorder risk variant is associated with limbic gray matter deficits in healthy subjects and major depression. Neuropsychopharmacology. 2015; 40(11): 25102516.Google Scholar
Kim, BJ, Zaveri, HP, Shchelochkov, OA, et al. An allelic series of mice reveals a role for RERE in the development of multiple organs affected in chromosome 1p36 deletions. Plos One. 2013; 8(2): e57460.Google Scholar
Kakeda, S, Watanabe, K, Katsuki, A, et al. Genetic effects on white matter integrity in drug-naive patients with major depressive disorder: A diffusion tensor imaging study of 17 genetic loci associated with depressive symptoms. Neuropsych Dis Treat. 2019; 15: 375383.Google Scholar
Han, KM, Won, E, Kang, J, et al. TESC gene-regulating genetic variant (rs7294919) affects hippocampal subfield volumes and parahippocampal cingulum white matter integrity in major depressive disorder. J Psychiat Res. 2017; 93: 2029.CrossRefGoogle ScholarPubMed
Cui, LL, Gong, XH, Tang, YQ, et al. Relationship between the LHPP gene polymorphism and resting-state brain activity in major depressive disorder. Neural Plasticity. 2016: 9162590.Google Scholar
Ota, M, Hori, H, Sato, N, et al. Effects of ankyrin 3 gene risk variants on brain structures in patients with bipolar disorder and healthy subjects. Psychiatry Clin Neurosci. 2016; 70(11): 498506.CrossRefGoogle ScholarPubMed
Lippard, ETC, Jensen, KP, Wang, F, et al. Genetic variation of ANK3 is associated with lower white matter structural integrity in bipolar disorder. Mol Psychiatry. 2017; 22(9): 1225.CrossRefGoogle ScholarPubMed
Delvecchio, G, Dima, D, Frangou, S. The effect of ANK3 bipolar-risk polymorphisms on the working memory circuitry differs between loci and according to risk-status for bipolar disorder. Am J Med Genet B Neuropsychiatr Genet. 2015; 168B(3): 188196.Google Scholar
Tesli, M, Egeland, R, Sonderby, IE, et al. No evidence for association between bipolar disorder risk gene variants and brain structural phenotypes. J Affect Disord. 2013; 151(1): 291297.Google Scholar
Falls, DL. Neuregulins: Functions, forms, and signaling strategies. Exp Cell Res. 2003; 284(1): 1430.Google Scholar
Cannon, DM, Walshe, M, Dempster, E, et al. The association of white matter volume in psychotic disorders with genotypic variation in NRG1, MOG and CNP: A voxel-based analysis in affected individuals and their unaffected relatives. Transl Psychiatry. 2012; 2: e167.Google Scholar
Mechelli, A, Prata, DP, Fu, CH, et al. The effects of neuregulin1 on brain function in controls and patients with schizophrenia and bipolar disorder. Neuroimage. 2008; 42(2): 817826.Google Scholar
Gomez-Ospina, N, Tsuruta, F, Barreto-Chang, O, Hu, L, Dolmetsch, R. The C terminus of the L-type voltage-gated calcium channel Ca(V)1.2 encodes a transcription factor. Cell. 2006; 127(3): 591606.Google Scholar
Green, EK, Grozeva, D, Jones, I, et al. The bipolar disorder risk allele at CACNA1C also confers risk of recurrent major depression and of schizophrenia. Mol Psychiatr. 2010; 15(10): 10161022.Google Scholar
Krug, A, Nieratschker, V, Markov, V, et al. Effect of CACNA1C rs1006737 on neural correlates of verbal fluency in healthy individuals. Neuroimage. 2010; 49(2): 18311836.Google Scholar
Thimm, M, Kircher, T, Kellermann, T, et al. Effects of a CACNA1C genotype on attention networks in healthy individuals. Psychological Medicine. 2011; 41(7): 15511561.CrossRefGoogle ScholarPubMed
Backes, H, Dietsche, B, Nagels, A, et al. Genetic variation in CACNA1C affects neural processing in major depression. J Psychiat Res. 2014; 53: 3846.CrossRefGoogle ScholarPubMed
Soeiro-de-Souza, MG, Lafer, B, Moreno, RA, et al. The CACNA1C risk allele rs1006737 is associated with age-related prefrontal cortical thinning in bipolar I disorder. Transl Psychiatry. 2017; 7(4): e1086.CrossRefGoogle ScholarPubMed
Perrier, E, Pompei, F, Ruberto, G, et al. Initial evidence for the role of CACNA1 C on subcortical brain morphology in patients with bipolar disorder. Eur Psychiatry. 2011; 26(3): 135137.Google Scholar
Tesli, M, Skatun, KC, Ousdal, OT, et al. CACNA1C risk variant and amygdala activity in bipolar disorder, schizophrenia and healthy controls. PLoS One. 2013; 8(2): e56970.Google Scholar
Dima, D, Jogia, J, Collier, D, et al. Independent modulation of engagement and connectivity of the facial network during affect processing by CACNA1C and ANK3 risk genes for bipolar disorder. JAMA Psychiatry. 2013; 70(12): 13031311.Google Scholar
Jogia, J, Ruberto, G, Lelli-Chiesa, G, et al. The impact of the CACNA1C gene polymorphism on frontolimbic function in bipolar disorder. Mol Psychiatry. 2011; 16(11): 10701071.Google Scholar
Whalley, HC, McKirdy, J, Romaniuk, L, et al. Functional imaging of emotional memory in bipolar disorder and schizophrenia. Bipolar Disord. 2009; 11(8): 840856.Google Scholar
Bigos, KL, Mattay, VS, Callicott, JH, et al. Genetic variation in CACNA1C affects brain circuitries related to mental illness. Arch Gen Psychiatry. 2010; 67(9): 939945.Google Scholar
Tecelao, D, Mendes, A, Martins, D, et al. The effect of psychosis associated CACNA1C, and its epistasis with ZNF804A, on brain function. Genes Brain Behav. 2019; 18(4): e12510.Google Scholar
Mallas, E, Carletti, F, Chaddock, CA, et al. The impact of CACNA1C gene, and its epistasis with ZNF804A, on white matter microstructure in health, schizophrenia and bipolar disorder(1). Genes Brain Behav. 2017; 16(4): 479488.Google Scholar
Soeiro-de-Souza, MG, Otaduy, MC, Dias, CZ, et al.The impact of the CACNA1C risk allele on limbic structures and facial emotions recognition in bipolar disorder subjects and healthy controls. J Affect Disord. 2012; 141(1): 94101.Google Scholar
Wolf, C, Mohr, H, Schneider-Axmann, T, et al. CACNA1 C genotype explains interindividual differences in amygdala volume among patients with schizophrenia. Eur Arch Psychiatry Clin Neurosci. 2014; 264(2): 93102.Google Scholar
Radua, J, Surguladze, SA, Marshall, N, et al. The impact of CACNA1C allelic variation on effective connectivity during emotional processing in bipolar disorder. Mol Psychiatry. 2013; 18(5): 526527.Google Scholar
Byrne, EM, Carrillo-Roa, T, Penninx, BW, et al. Applying polygenic risk scores to postpartum depression. Arch Womens Ment Health. 2014; 17(6): 519528.Google Scholar
Levine, ME, Crimmins, EM, Prescott, CA, et al. A polygenic risk score associated with measures of depressive symptoms among older adults. Biodemogr Soc Biol. 2014; 60(2): 199211.Google Scholar
Mullins, N, Power, RA, Fisher, HL, et al. Polygenic interactions with environmental adversity in the aetiology of major depressive disorder. Psychological Medicine. 2016; 46(4): 759770.Google Scholar
Holmes, AJ, Lee, PH, Hollinshead, MO, et al. Individual differences in amygdala-medial prefrontal anatomy link negative affect, impaired social functioning, and polygenic depression risk. Journal of Neuroscience. 2012; 32(50): 1808718100.Google Scholar
Whalley, HC, Sprooten, E, Hackett, S, et al. Polygenic risk and white matter integrity in individuals at high risk of mood disorder. Biol Psychiat. 2013; 74(4): 280286.Google Scholar
Yuksel, D, Dietsche, B, Forstner, AJ, et al. Polygenic risk for depression and the neural correlates of working memory in healthy subjects. Prog Neuro-Psychoph. 2017; 79: 6776.Google Scholar
Reus, LM, Shen, X, Gibson, J, et al. Association of polygenic risk for major psychiatric illness with subcortical volumes and white matter integrity in UK Biobank. Sci Rep-Uk. 2017; 7: 42140.Google Scholar
Wigmore, EM, Clarke, TK, Howard, DM, et al. Do regional brain volumes and major depressive disorder share genetic architecture? A study of Generation Scotland (n=19762), UK Biobank (n=24048) and the English Longitudinal Study of Ageing (n=5766). Transl Psychiatry. 2017 August 15; 7(8): e1205.Google Scholar
Wang, T, Zhang, X, Li, A, et al. Polygenic risk for five psychiatric disorders and cross-disorder and disorder-specific neural connectivity in two independent populations. NeuroImage Clinical. 2017; 14: 441449.Google Scholar
Tesli, M, Kauppi, K, Bettella, F, et al. Altered brain activation during emotional face processing in relation to both diagnosis and polygenic risk of bipolar disorder. PLoS One. 2015; 10(7): e0134202.Google Scholar
Dima, D, Breen, G. Polygenic risk scores in imaging genetics: Usefulness and applications. J Psychopharmacol. 2015; 29(8): 867871.Google Scholar
Whalley, HC, Papmeyer, M, Sprooten, E, et al. The influence of polygenic risk for bipolar disorder on neural activation assessed using fMRI. Transl Psychiatry. 2012; 2: e130.Google Scholar
Caseras, X, Tansey, KE, Foley, S, Linden, D. Association between genetic risk scoring for schizophrenia and bipolar disorder with regional subcortical volumes. Transl Psychiatry. 2015; 5: e692.Google Scholar
Wang, T, Zhang, X, Li, A, et al. Polygenic risk for five psychiatric disorders and cross-disorder and disorder-specific neural connectivity in two independent populations. Neuroimage Clin. 2017; 14: 441449.Google Scholar
Doan, NT, Kaufmann, T, Bettella, F, et al. Distinct multivariate brain morphological patterns and their added predictive value with cognitive and polygenic risk scores in mental disorders. Neuroimage Clin. 2017; 15: 719731.CrossRefGoogle ScholarPubMed
Whalley, HC, Sprooten, E, Hackett, S, et al. Polygenic risk and white matter integrity in individuals at high risk of mood disorder. Biol Psychiatry. 2013; 74(4): 280286.Google Scholar
Reus, LM, Shen, X, Gibson, J, et al. Association of polygenic risk for major psychiatric illness with subcortical volumes and white matter integrity in UK Biobank. Sci Rep. 2017; 7: 42140.Google Scholar
Sprooten, E, Fleming, KM, Thomson, PA, et al. White matter integrity as an intermediate phenotype: Exploratory genome-wide association analysis in individuals at high risk of bipolar disorder. Psychiatry Res. 2013; 206(2–3): 223231.CrossRefGoogle ScholarPubMed
Serretti, A, Cusin, C, Benedetti, F, et al. Insomnia improvement during antidepressant treatment and CLOCK gene polymorphism. Am J Med Genet B Neuropsychiatr Genet. 2005; 137B(1): 3639.Google Scholar
Zai, CC, de Luca, V, Strauss, J, et al. Genetic factors and suicidal behavior. In: Dwivedi, Y, editor. The Neurobiological Basis of Suicide. Frontiers in Neuroscience. Boca Raton (FL) Florida, US: CRC Press/Taylor & Francis; 2012, p. 213.Google Scholar
Kenna, GA, Roder-Hanna, N, Leggio, L, et al. Association of the 5-HTT gene-linked promoter region (5-HTTLPR) polymorphism with psychiatric disorders: review of psychopathology and pharmacotherapy. Pharmgenomics Pers Med. 2012; 5: 1935.Google Scholar
Smeraldi, E, Benedetti, F, Zanardi, R. Serotonin transporter promoter genotype and illness recurrence in mood disorders. Eur Neuropsychopharmacol. 2002; 12(1): 7375.Google Scholar
Serretti, A, Kato, M, De Ronchi, D, Kinoshita, T. Meta-analysis of serotonin transporter gene promoter polymorphism (5-HTTLPR) association with selective serotonin reuptake inhibitor efficacy in depressed patients. Mol Psychiatry. 2007; 12(3): 247257.CrossRefGoogle ScholarPubMed
Parra-Uribe, I, Blasco-Fontecilla, H, Garcia-Pares, G, et al. Risk of re-attempts and suicide death after a suicide attempt: A survival analysis. BMC Psychiatry. 2017; 17(1): 163.Google Scholar
Russ, MJ, Lachman, HM, Kashdan, T, Saito, T, Bajmakovic-Kacila, S. Analysis of catechol-O-methyltransferase and 5-hydroxytryptamine transporter polymorphisms in patients at risk for suicide. Psychiatry Res. 2000; 93(1): 7378.Google Scholar
Murphy, SE, Norbury, R, Godlewska, BR, et al. The effect of the serotonin transporter polymorphism (5-HTTLPR) on amygdala function: A meta-analysis. Mol Psychiatry. 2013; 18(4): 512520.Google Scholar
Munafo, MR, Brown, SM, Hariri, AR. Serotonin transporter (5-HTTLPR) genotype and amygdala activation: A meta-analysis. Biol Psychiatry. 2008; 63(9): 852857.CrossRefGoogle ScholarPubMed
Pezawas, L, Meyer-Lindenberg, A, Drabant, EM, et al. 5-HTTLPR polymorphism impacts human cingulate-amygdala interactions: A genetic susceptibility mechanism for depression. Nat Neurosci. 2005; 8(6): 828834.Google Scholar
Jonassen, R, Landro, NI. Serotonin transporter polymorphisms (5-HTTLPR) in emotion processing: Implications from current neurobiology. Prog Neurobiol. 2014; 117: 4153.Google Scholar
Frodl, T, Zill, P, Baghai, T, et al. Reduced hippocampal volumes associated with the long variant of the tri- and diallelic serotonin transporter polymorphism in major depression. American Journal of Medical Genetics Part B, Neuropsychiatric Genetics: The Official Publication of the International Society of Psychiatric Genetics. 2008; 147B(7): 10031007.Google Scholar
Frodl, T, Meisenzahl, EM, Zill, P, et al. Reduced hippocampal volumes associated with the long variant of the serotonin transporter polymorphism in major depression. Arch Gen Psychiat. 2004; 61(2): 177183.CrossRefGoogle Scholar
Taylor, WD, Steffens, DC, Payne, ME, et al. Influence of serotonin transporter promoter region polymorphisms on hippocampal volumes in late-life depression. Arch Gen Psychiat. 2005; 62(5): 537544.Google Scholar
Eker, MC, Kitis, O, Okur, H, et al. Smaller hippocampus volume is associated with short variant of 5-HTTLPR polymorphism in medication-free major depressive disorder patients. Neuropsychobiology. 2011; 63(1): 2228.CrossRefGoogle ScholarPubMed
Hickie, IB, Naismith, SL, Ward, PB, et al. Serotonin transporter gene status predicts caudate nucleus but not amygdala or hippocampal volumes in older persons with major depression. J Affect Disord. 2007; 98(1–2): 137142.CrossRefGoogle ScholarPubMed
Jaworska, N, MacMaster, FP, Foster, J, Ramasubbu, R. The influence of 5-HTTLPR and Val66 Met polymorphisms on cortical thickness and volume in limbic and paralimbic regions in depression: A preliminary study. BMC Psychiatry. 2016; 16: 61.Google Scholar
Ahdidan, J, Foldager, L, Rosenberg, R, et al. Hippocampal volume and serotonin transporter polymorphism in major depressive disorder. Acta Neuropsychiatr. 2013; 25(4): 206214.Google Scholar
Cole, J, Weinberger, DR, Mattay, VS, et al. No effect of 5HTTLPR or BDNF Val66 Met polymorphism on hippocampal morphology in major depression. Genes, Brain, and Behavior. 2011; 10(7): 756764.CrossRefGoogle ScholarPubMed
Hickie, IB, Naismith, SL, Ward, PB, et al. Serotonin transporter gene status predicts caudate nucleus but not amygdala or hippocampal volumes in older persons with major depression. J Affect Disorders. 2007; 98(1–2): 137142.Google Scholar
Taylor, WD, Macfall, JR, Payne, ME, et al. Orbitofrontal cortex volume in late life depression: Influence of hyperintense lesions and genetic polymorphisms. Psychological Medicine. 2007; 37(12): 17631773.Google Scholar
Costafreda, SG, McCann, P, Saker, P, et al. Modulation of amygdala response and connectivity in depression by serotonin transporter polymorphism and diagnosis. J Affect Disorders. 2013; 150(1): 96103.Google Scholar
Dannlowski, U, Ohrmann, P, Bauer, J, et al. 5-HTTLPR biases amygdala activity in response to masked facial expressions in major depression. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2008; 33(2): 418424.Google Scholar
Friedel, E, Schlagenhauf, F, Sterzer, P, et al. 5-HTT genotype effect on prefrontal-amygdala coupling differs between major depression and controls. Psychopharmacology. 2009; 205(2): 261271.Google Scholar
Benedetti, F, Bollettini, I, Poletti, S, et al. White matter microstructure in bipolar disorder is influenced by the serotonin transporter gene polymorphism 5-HTTLPR. Genes Brain Behav. 2015; 14(3): 238250.CrossRefGoogle ScholarPubMed
Frodl, T, Reinhold, E, Koutsouleris, N, et al. Childhood stress, serotonin transporter gene and brain structures in major depression. Neuropsychopharmacology: Official publication of the American College of Neuropsychopharmacology. 2010; 35(6): 13831390.Google Scholar
Alexopoulos, GS, Murphy, CF, Gunning-Dixon, FM, et al. Serotonin transporter polymorphisms, microstructural white matter abnormalities and remission of geriatric depression. J Affect Disorders. 2009; 119(1–3): 132141.Google Scholar
Parsey, RV, Olvet, DM, Oquendo, MA, et al. Higher 5-HT1A receptor binding potential during a major depressive episode predicts poor treatment response: Preliminary data from a naturalistic study. Neuropsychopharmacology. 2006; 31(8): 17451749.Google Scholar
Lemonde, S, Turecki, G, Bakish, D, et al. Impaired repression at a 5-hydroxytryptamine 1 A receptor gene polymorphism associated with major depression and suicide. The Journal of Neuroscience. 2003; 23(25): 87888799.Google Scholar
Vai, B, Riberto, M, Ghiglino, D, et al. A 5-HT1Areceptor promoter polymorphism influences fronto-limbic functional connectivity and depression severity in bipolar disorder. Psychiatry Res Neuroimaging. 2017; 270: 17.Google Scholar
Bilder, RM, Volavka, J, Lachman, HM, Grace, AA. The catechol-O-methyltransferase polymorphism: Relations to the tonic-phasic dopamine hypothesis and neuropsychiatric phenotypes. Neuropsychopharmacology. 2004; 29(11): 19431961.Google Scholar
Benedetti, F, Dallaspezia, S, Colombo, C, et al. Effect of catechol-O-methyltransferase Val(108/158)Met polymorphism on antidepressant efficacy of fluvoxamine. Eur Psychiatry. 2010; 25(8): 476478.CrossRefGoogle ScholarPubMed
Benedetti, F, Barbini, B, Bernasconi, A, et al. Acute antidepressant response to sleep deprivation combined with light therapy is influenced by the catechol-O-methyltransferase Val(108/158)Met polymorphism. J Affect Disord. 2010; 121(1–2): 6872.Google Scholar
Benedetti, F, Dallaspezia, S, Locatelli, C, et al. Recurrence of bipolar mania is associated with catechol-O-methyltransferase Val(108/158)Met polymorphism. J Affect Disord. 2011; 132(1–2): 293296.Google Scholar
Benedetti, F, Dallaspezia, S, Colombo, C, et al. Association between catechol-O-methyltransferase Val(108/158)Met polymorphism and psychotic features of bipolar disorder. J Affect Disord. 2010; 125(1–3): 341344.Google Scholar
Papolos, DF, Veit, S, Faedda, GL, Saito, T, Lachman, HM. Ultra-ultra rapid cycling bipolar disorder is associated with the low activity catecholamine-O-methyltransferase allele. Mol Psychiatry. 1998; 3(4): 346349.CrossRefGoogle ScholarPubMed
Seok, JH, Choi, S, Lim, HK, et al. Effect of the COMT val158 met polymorphism on white matter connectivity in patients with major depressive disorder. Neurosci Lett. 2013; 545: 3539.CrossRefGoogle Scholar
Hayashi, K, Yoshimura, R, Kakeda, S, et al. COMT Val158 Met, but not BDNF Val66 Met, is associated with white matter abnormalities of the temporal lobe in patients with first-episode, treatment-naive major depressive disorder: A diffusion tensor imaging study. Neuropsych Dis Treat. 2014; 10: 11831190.Google Scholar
Watanabe, K, Kakeda, S, Yoshimura, R, et al. Relationship between the catechol-O-methyl transferase Va1108/158 Met genotype and brain volume in treatment-naive major depressive disorder: Voxel-based morphometry analysis. Psychiat Res-Neuroim. 2015; 233(3): 481487.Google Scholar
Opmeer, EM, Kortekaas, R, van Tol, MJ, et al. Influence of COMT val158 met genotype on the depressed brain during emotional processing and working memory. PLoS One. 2013; 8(9): e73290.Google Scholar
Vai, B, Riberto, M, Poletti, S, et al. Catechol-O-methyltransferase Val (108/158) Met polymorphism affects fronto-limbic connectivity during emotional processing in bipolar disorder. European Psychiatry. 2017; 41: 5359.Google Scholar
Miskowiak, KW, Kjaerstad, HL, Stottrup, MM, et al. The catechol-O-methyltransferase (COMT) Val158 Met genotype modulates working memory-related dorsolateral prefrontal response and performance in bipolar disorder. Bipolar Disord. 2017; 19(3): 214224.CrossRefGoogle Scholar
Hempstead, BL. Brain-derived neurotrophic factor: Three ligands, many actions. Trans Am Clin Climatol Assoc. 2015; 126: 919.Google ScholarPubMed
Notaras, M, Hill, R, van den Buuse, M. The BDNF gene Val66 Met polymorphism as a modifier of psychiatric disorder susceptibility: Progress and controversy. Mol Psychiatry. 2015; 20(8): 916930.Google Scholar
Ide, S, Kakeda, S, Watanabe, K, et al. Relationship between a BDNF gene polymorphism and the brain volume in treatment-naive patients with major depressive disorder: A VBM analysis of brain MRI. Psychiatry Res. 2015; 233(2): 120124.Google Scholar
Legge, RM, Sendi, S, Cole, JH, et al. Modulatory effects of brain-derived neurotrophic factor Val66 Met polymorphism on prefrontal regions in major depressive disorder. Br J Psychiatry. 2015; 206(5): 379384.Google Scholar
Frodl, T, Schule, C, Schmitt, G, et al. Association of the brain-derived neurotrophic factor Val66 Met polymorphism with reduced hippocampal volumes in major depression. Arch Gen Psychiatry. 2007; 64(4): 410416.CrossRefGoogle Scholar
Carballedo, A, Morris, D, Zill, P, et al. Brain-derived neurotrophic factor Val66 Met polymorphism and early life adversity affect hippocampal volume. Am J Med Genet B Neuropsychiatr Genet. 2013; 162B(2): 183190.Google Scholar
Youssef, MM, Underwood, MD, Huang, YY, et al. Association of BDNF Val66 Met polymorphism and brain BDNF levels with major depression and suicide. Int J Neuropsychopharmacol. 2018; 21(6): 528538.Google Scholar
Alexopoulos, GS, Glatt, CE, Hoptman, MJ, et al. BDNF val66 met polymorphism, white matter abnormalities and remission of geriatric depression. J Affect Disord. 2010; 125(1–3): 262268.Google Scholar
Han, KM, Choi, S, Kim, A, et al. The effects of 5-HTTLPR and BDNF Val66 Met polymorphisms on neurostructural changes in major depressive disorder. Psychiatry Res Neuroimaging. 2018; 273: 2534.Google Scholar
Tatham, EL, Hall, GBC, Clark, D, Foster, J, Ramasubbu, R. The 5-HTTLPR and BDNF polymorphisms moderate the association between uncinate fasciculus connectivity and antidepressants treatment response in major depression. European Archives of Psychiatry and Clinical Neuroscience. 2017; 267(2): 135147.CrossRefGoogle ScholarPubMed
Yin, YY, Hou, ZH, Wang, XQ, Sui, YX, Yuan, YG. The BDNF Val66 Met polymorphism, resting-state hippocampal functional connectivity and cognitive deficits in acute late-onset depression. J Affect Disorders. 2015; 183: 2230.Google Scholar
Opmeer, EM, Kortekaas, R, van Tol, MJ, et al. Influence of COMT val158 met genotype on the depressed brain during emotional processing and working memory. Plos One. 2013 September 12; 8(9): e73290.Google Scholar
Gonul, AS, Kitis, O, Eker, MC, et al. Association of the brain-derived neurotrophic factor Val66 Met polymorphism with hippocampus volumes in drug-free depressed patients. World J Biol Psychia. 2011; 12(2): 110118.Google Scholar
Kanellopoulos, D, Gunning, FM, Morimoto, SS, et al. Hippocampal volumes and the brain-derived neurotrophic factor val66 met polymorphism in geriatric major depression. Am J Geriatr Psychiatry. 2011; 19(1): 1322.CrossRefGoogle Scholar
Cao, B, Bauer, IE, Sharma, AN, et al. Reduced hippocampus volume and memory performance in bipolar disorder patients carrying the BDNF val66 met met allele. J Affect Disord. 2016; 198: 198205.Google Scholar
Chepenik, LG, Fredericks, C, Papademetris, X, et al. Effects of the brain-derived neurotrophic growth factor val66 met variation on hippocampus morphology in bipolar disorder. Neuropsychopharmacology. 2009; 34(4): 944951.CrossRefGoogle Scholar
Matsuo, K, Walss-Bass, C, Nery, FG, et al. Neuronal correlates of brain-derived neurotrophic factor Val66 Met polymorphism and morphometric abnormalities in bipolar disorder. Neuropsychopharmacology. 2009; 34(8): 19041913.Google Scholar
Mirakhur, A, Moorhead, TW, Stanfield, AC, et al. Changes in gyrification over 4 years in bipolar disorder and their association with the brain-derived neurotrophic factor valine(66) methionine variant. Biol Psychiatry. 2009; 66(3): 293297.Google Scholar
Zeni, CP, Mwangi, B, Cao, B, et al. Interaction between BDNF rs6265 Met allele and low family cohesion is associated with smaller left hippocampal volume in pediatric bipolar disorder. J Affect Disord. 2016; 189: 9497.CrossRefGoogle ScholarPubMed
Mandolini, GM, Lazzaretti, M, Pigoni, A, et al. The impact of BDNF Val66 Met polymorphism on cognition in bipolar disorder: A review: Special section on “translational and neuroscience studies in affective disorders” section editor, Maria Nobile MD, PhD. This section of JAD focuses on the relevance of translational and neuroscience studies in providing a better understanding of the neural basis of affective disorders. The main aim is to briefly summaries relevant research findings in clinical neuroscience with particular regards to specific innovative topics in mood and anxiety disorders. J Affect Disord. 2019; 243: 552558.Google Scholar
Fernandes, BS, Molendijk, ML, Kohler, CA, et al. Peripheral brain-derived neurotrophic factor (BDNF) as a biomarker in bipolar disorder: a meta-analysis of 52 studies. BMC Med. 2015; 13: 289.Google Scholar
Chen, J, Xu, Y, Zhang, J, et al. Genotypic association of the DAOA gene with resting-state brain activity in major depression. Mol Neurobiol. 2012; 46(2): 361373.CrossRefGoogle ScholarPubMed
Choi, S, Han, KM, Kang, J, et al. Effects of a polymorphism of the neuronal amino acid transporter SLC6A15 gene on structural integrity of white matter tracts in major depressive disorder. Plos One. 2016 October 10; 11(10): e0164301.Google Scholar
Wang, LJ, Liu, ZF, Cao, XH, et al. A combined study of SLC6A15 gene polymorphism and the resting-state functional magnetic resonance imaging in first-episode drug-naive major depressive disorder. Genet Test Mol Bioma. 2017; 21(9): 523530.CrossRefGoogle ScholarPubMed
Poletti, S, Riberto, M, Vai, B, et al. A glutamate transporter EAAT1 gene variant influences amygdala functional connectivity in bipolar disorder. Journal of Molecular Neuroscience. 2018; 65(4): 536545.CrossRefGoogle ScholarPubMed
Poletti, S, Bollettini, I, Lorenzi, C, et al. White matter microstructure in bipolar disorder is influenced by the interaction between a glutamate transporter EAAT1 gene variant and early stress. Molecular Neurobiology. 2018: 19.Google Scholar
Benedetti, F, Poletti, S, Locatelli, C, et al. A Homer 1 gene variant influences brain structure and function, lithium effects on white matter, and antidepressant response in bipolar disorder: A multimodal genetic imaging study. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2018; 81: 8895.CrossRefGoogle Scholar
Benedetti, F, Poletti, S, Locatelli, C, et al. A Homer 1 gene variant influences brain structure and function, lithium effects on white matter, and antidepressant response in bipolar disorder: A multimodal genetic imaging study. Prog Neuropsychopharmacol Biol Psychiatry. 2018; 81: 8895.CrossRefGoogle Scholar
Bollettini, I, Melloni, EMT, Aggio, V, et al. Clock genes associate with white matter integrity in depressed bipolar patients. Chronobiology International. 2017; 34(2): 212224.Google Scholar
Poletti, S, Aggio, V, Bollettini, I, et al. SREBF-2 polymorphism influences white matter microstructure in bipolar disorder. Psychiatry Research: Neuroimaging. 2016; 257: 3946.Google Scholar
Grimes, CA, Jope, RS. CREB DNA binding activity is inhibited by glycogen synthase kinase-3 beta and facilitated by lithium. J Neurochem. 2001; 78(6): 12191232.Google Scholar
Inkster, B, Nichols, TE, Saemann, PG, et al. Association of GSK3beta polymorphisms with brain structural changes in major depressive disorder. Arch Gen Psychiatry. 2009; 66(7): 721728.Google Scholar
Inkster, B, Simmons, A, Cole, JH, et al. Unravelling the GSK3 beta-related genotypic interaction network influencing hippocampal volume in recurrent major depressive disorder. Psychiatric Genetics. 2018; 28(5): 7784.Google Scholar
Liu, Z, Guo, H, Cao, XH, et al. A combined study of GSK3 beta polymorphisms and brain network topological metrics in major depressive disorder. Psychiat Res-Neuroim. 2014; 223(3): 210217.Google Scholar
Inkster, B, Nichols, TE, Saemann, PG, et al. Pathway-based approaches to imaging genetics association studies: Wnt signaling, GSK3beta substrates and major depression. Neuroimage. 2010; 53(3): 908917.Google Scholar
Benedetti, F, Serretti, A, Colombo, C, et al. A glycogen synthase kinase 3-beta promoter gene single nucleotide polymorphism is associated with age at onset and response to total sleep deprivation in bipolar depression. Neurosci Lett. 2004; 368(2): 123126.Google Scholar
Benedetti, F, Bollettini, I, Barberi, I, et al. Lithium and GSK3-β promoter gene variants influence white matter microstructure in bipolar disorder. Neuropsychopharmacology. 2013; 38(2): 313.Google Scholar
Horstmann, S, Lucae, S, Menke, A, et al. Polymorphisms in GRIK4, HTR2A, and FKBP5 show interactive effects in predicting remission to antidepressant treatment. Neuropsychopharmacology. 2010; 35(3): 727740.Google Scholar
Binder, EB, Salyakina, D, Lichtner, P, et al. Polymorphisms in FKBP5 are associated with increased recurrence of depressive episodes and rapid response to antidepressant treatment. Nat Genet. 2004; 36(12): 13191325.Google Scholar
Pace, TW, Miller, AH. Cytokines and glucocorticoid receptor signaling. Relevance to major depression. Ann N Y Acad Sci. 2009; 1179: 86105.Google Scholar
Cordova-Palomera, A, de Reus, MA, Fatjo-Vilas, M, et al. FKBP5 modulates the hippocampal connectivity deficits in depression: A study in twins. Brain Imaging Behav. 2017; 11(1): 6275.Google Scholar
Tozzi, L, Carballedo, A, Wetterling, F, et al. Single-nucleotide polymorphism of the FKBP5 gene and childhood maltreatment as predictors of structural changes in brain areas involved in emotional processing in depression. Neuropsychopharmacology. 2016; 41(2): 487497.CrossRefGoogle ScholarPubMed
Han, KM, Won, E, Sim, Y, et al. Influence of FKBP5 polymorphism and DNA methylation on structural changes of the brain in major depressive disorder. Sci Rep-Uk. 2017; 7.Google Scholar
Tozzi, L, Doolin, K, Farrel, C, et al. Functional magnetic resonance imaging correlates of emotion recognition and voluntary attentional regulation in depression: A generalized psycho-physiological interaction study. J Affect Disorders. 2017; 208: 535544.Google Scholar
Qiu, AQ, Taylor, WD, Zhao, Z, et al. APOE related hippocampal shape alteration in geriatric depression. Neuroimage. 2009; 44(3): 620626.Google Scholar
Kim, DH, Payne, ME, Levy, RM, MacFall, JR, Steffens, DC. APOE genotype and hippocampal volume change in geriatric depression. Biol Psychiat. 2002; 51(5): 426429.CrossRefGoogle ScholarPubMed
Sachs-Ericsson, N, Sawyer, K, Corsentino, E, Collins, N, Steffens, DC. The moderating effect of the APOE epsilon 4 allele on the relationship between hippocampal volume and cognitive decline in older depressed patients. Am J Geriat Psychiat. 2011; 19(1): 2332.Google Scholar
Yuan, YG, Zhang, ZJ, Bai, F, et al. Genetic variation in apolipoprotein E alters regional gray matter volumes in remitted late-onset depression. J Affect Disorders. 2010; 121(3): 273277.Google Scholar
Shu, H, Yuan, YG, Xie, CM, et al. Imbalanced hippocampal functional networks associated with remitted geriatric depression and apolipoprotein E epsilon 4 allele in nondemented elderly: A preliminary study. J Affect Disorders. 2014; 164: 513.Google Scholar
Wu, D, Yuan, YG, Ba, F, et al. Abnormal functional connectivity of the default mode network in remitted late-onset depression. J Affect Disorders. 2013; 147(1–3): 277287.Google Scholar
Chang, KJ, Hong, CH, Lee, KS, et al. Differential effects of white matter hyperintensity on geriatric depressive symptoms according to APOE-epsilon 4 status. J Affect Disorders. 2015; 188: 2834.Google Scholar
Lavretsky, H, Lesser, IM, Wohl, M, et al. Apolipoprotein-E and white matter hyperintensities in late-life depression. Am J Geriat Psychiat. 2000; 8(3): 257261.Google Scholar
Turecki, G, Ota, VK, Belangero, SI, Jackowski, A, Kaufman, J. Early life adversity, genomic plasticity, and psychopathology. Lancet Psychiatry. 2014; 1(6): 461466.Google Scholar
Lutz, PE, Turecki, G. DNA methylation and childhood maltreatment: From animal models to human studies. Neuroscience. 2014; 264: 142156.Google Scholar
Szyf, M. The early life social environment and DNA methylation: DNA methylation mediating the long-term impact of social environments early in life. Epigenetics. 2011; 6(8): 971978.Google Scholar
Caspi, A, Hariri, AR, Holmes, A, Uher, R, Moffitt, TE. Genetic sensitivity to the environment: The case of the serotonin transporter gene and its implications for studying complex diseases and traits. Am J Psychiatry. 2010; 167(5): 509527.Google Scholar
Philibert, RA, Sandhu, H, Hollenbeck, N, et al. The relationship of 5HTT (SLC6A4) methylation and genotype on mRNA expression and liability to major depression and alcohol dependence in subjects from the Iowa adoption studies. Am J Med Genet B Neuropsychiatr Genet. 2008; 147B(5): 543549.Google Scholar
Okada, S, Morinobu, S, Fuchikami, M, et al. The potential of SLC6A4 gene methylation analysis for the diagnosis and treatment of major depression. J Psychiatr Res. 2014; 53: 4753.Google Scholar
Domschke, K, Tidow, N, Schwarte, K, et al. Serotonin transporter gene hypomethylation predicts impaired antidepressant treatment response. Int J Neuropsychopharmacol. 2014; 17(8): 11671176.Google Scholar
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): 159165.Google Scholar
Kang, HJ, Kim, JM, Stewart, R, et al. Association of SLC6A4 methylation with early adversity, characteristics and outcomes in depression. Prog Neuropsychopharmacol Biol Psychiatry. 2013; 44: 2328.Google Scholar
Sugawara, H, Iwamoto, K, Bundo, M, et al. Hypermethylation of serotonin transporter gene in bipolar disorder detected by epigenome analysis of discordant monozygotic twins. Transl Psychiatry. 2011; 1: e24.CrossRefGoogle ScholarPubMed
Won, E, Choi, S, Kang, J, et al. Association between reduced white matter integrity in the corpus callosum and serotonin transporter gene DNA methylation in medication-naive patients with major depressive disorder. Transl Psychiatry. 2016; 6(8): e866.CrossRefGoogle ScholarPubMed
Booij, L, Szyf, M, Carballedo, A, et al. DNA methylation of the serotonin transporter gene in peripheral cells and stress-related changes in hippocampal volume: A study in depressed patients and healthy controls. PLoS One. 2015; 10(3): e0119061.Google Scholar
Kaer, K, Speek, M. Retroelements in human disease. Gene. 2013; 518(2): 231241.Google Scholar
Schneider, I, Kugel, H, Redlich, R, et al. Association of serotonin transporter gene AluJb methylation with major depression, amygdala responsiveness, 5-HTTLPR/rs25531 polymorphism, and stress. Neuropsychopharmacology. 2018; 43(6): 13081316.Google Scholar
Na, KS, Won, E, Kang, J, et al. Differential effect of COMT gene methylation on the prefrontal connectivity in subjects with depression versus healthy subjects. Neuropharmacology. 2018; 137: 5970.Google Scholar
Tyrka, AR, Ridout, KK, Parade, SH. Childhood adversity and epigenetic regulation of glucocorticoid signaling genes: Associations in children and adults. Dev Psychopathol. 2016; 28(4pt2): 13191331.Google Scholar
Tozzi, L, Farrell, C, Booij, L, et al. Epigenetic changes of FKBP5 as a link connecting genetic and environmental risk factors with structural and functional brain changes in major depression. Neuropsychopharmacology. 2018; 43(5): 11381145.Google Scholar
Han, KM, Won, E, Sim, Y, et al. Influence of FKBP5 polymorphism and DNA methylation on structural changes of the brain in major depressive disorder. Sci Rep. 2017; 7: 42621.Google Scholar
Na, KS, Chang, HS, Won, E, et al. Association between glucocorticoid receptor methylation and hippocampal subfields in major depressive disorder. PLoS One. 2014; 9(1): e85425.Google Scholar
Palma-Gudiel, H, Cordova-Palomera, A, Tornador, C, et al. Increased methylation at an unexplored glucocorticoid responsive element within exon 1D of NR3C1 gene is related to anxious-depressive disorders and decreased hippocampal connectivity. Eur Neuropsychopharmacol. 2018; 28(5): 579588.CrossRefGoogle ScholarPubMed
Fuchikami, M, Morinobu, S, Segawa, M, et al. DNA methylation profiles of the brain-derived neurotrophic factor (BDNF) gene as a potent diagnostic biomarker in major depression. PLoS One. 2011; 6(8): e23881.CrossRefGoogle ScholarPubMed
Tadic, A, Muller-Engling, L, Schlicht, KF, et al. Methylation of the promoter of brain-derived neurotrophic factor exon IV and antidepressant response in major depression. Mol Psychiatry. 2014; 19(3): 281283.Google Scholar
Kang, HJ, Kim, JM, Lee, JY, et al. BDNF promoter methylation and suicidal behavior in depressive patients. J Affect Disord. 2013; 151(2): 679685.CrossRefGoogle ScholarPubMed
Martinowich, K, Hattori, D, Wu, H, et al. DNA methylation-related chromatin remodeling in activity-dependent BDNF gene regulation. Science. 2003; 302(5646): 890893.Google Scholar
Choi, S, Han, KM, Won, E, et al. Association of brain-derived neurotrophic factor DNA methylation and reduced white matter integrity in the anterior corona radiata in major depression. J Affect Disord. 2015; 172: 7480.Google Scholar
Han, KM, Won, E, Kang, J, et al. TESC gene-regulating genetic variant (rs7294919) affects hippocampal subfield volumes and parahippocampal cingulum white matter integrity in major depressive disorder. J Psychiatr Res. 2017; 93: 2029.Google Scholar

References

Linden, DE. Neurofeedback and networks of depression. Dialogues Clin Neurosci. 2014; 16(1): 103112.Google Scholar
Esmail, S, Linden, DE. Emotion regulation networks and neurofeedback in depression. Cognitive Sciences. 2011; 6(2): 101.Google Scholar
Linden, DE. Brain Control: Developments in Therapy and Implications for Society. Springer; 2014.Google Scholar
Emmert, K, Kopel, R, Sulzer, J, et al. Meta-analysis of real-time fMRI neurofeedback studies using individual participant data: How is brain regulation mediated? Neuroimage. 2016; 124: 806812.CrossRefGoogle Scholar
Engen, HG, Kanske, P, Singer, T. The neural component-process architecture of endogenously generated emotion. Social Cognitive and Affective Neuroscience. 2016; 12(2): 197211.Google Scholar
Dörfel, D, Lamke, J-P, Hummel, F, et al. Common and differential neural networks of emotion regulation by detachment, reinterpretation, distraction, and expressive suppression: a comparative fMRI investigation. Neuroimage. 2014; 101: 298309.Google Scholar
Cohen, JR, Berkman, ET, Lieberman, MD. Intentional and incidental self-control in ventrolateral PFC. In: Donald, T. Stuss and Robert, T. Knight. editors. Principles of Frontal Lobe Function. 2nd edition. Oxford University Press; 2013, pp. 417–40.Google Scholar
Anderson, JS, Ferguson, MA, Lopez-Larson, M, Yurgelun-Todd, D. Connectivity gradients between the default mode and attention control networks. Brain Connectivity. 2011; 1(2): 147157.Google Scholar
Singh, KD, Fawcett, I. Transient and linearly graded deactivation of the human default-mode network by a visual detection task. Neuroimage. 2008; 41(1): 100112.CrossRefGoogle ScholarPubMed
Sridharan, D, Levitin, DJ, Menon, V. A critical role for the right fronto-insular cortex in switching between central-executive and default-mode networks. Proceedings of the National Academy of Sciences. 2008; 105(34): 1256912574.CrossRefGoogle ScholarPubMed
Scharnowski, F, Veit, R, Zopf, R, et al. Manipulating motor performance and memory through real-time fMRI neurofeedback. Biological Psychology. 2015; 108: 8597.CrossRefGoogle ScholarPubMed
Skottnik, L, Sorger, B, Kamp, T, Linden, D, Goebel, R. Success and failure of controlling the real‐time functional magnetic resonance imaging neurofeedback signal are reflected in the striatum. Brain and Behavior. 2019 March; 9(3): e01240.Google Scholar
Sitaram, R, Ros, T, Stoeckel, L, et al. Closed-loop brain training: The science of neurofeedback. Nature Reviews Neuroscience. 2017; 18(2): 86.Google Scholar
Mehler, DM, Sokunbi, MO, Habes, I, et al. Targeting the affective brain—a randomized controlled trial of real-time fMRI neurofeedback in patients with depression. Neuropsychopharmacology. 2018; 43(13): 2578.Google Scholar
Young, KD, Siegle, GJ, Zotev, V, et al. Randomized clinical trial of real-time fMRI amygdala neurofeedback for major depressive disorder: Effects on symptoms and autobiographical memory recall. American Journal of Psychiatry. 2017 August 1; 174(8): 748755.Google Scholar
Herwig, U, Kaffenberger, T, Jäncke, L, Brühl, AB. Self-related awareness and emotion regulation. NeuroImage. 2010; 50(2): 734741.Google Scholar
Beer, JS, Heerey, EA, Keltner, D, Scabini, D, Knight, RT. The regulatory function of self-conscious emotion: Insights from patients with orbitofrontal damage. Journal of Personality and Social Psychology. 2003; 85(4): 594.Google Scholar
Schooler, JW, Smallwood, J, Christoff, K, et al. Meta-awareness, perceptual decoupling and the wandering mind. Trends in Cognitive Sciences. 2011; 15(7): 319326.Google Scholar
Ernst, J, Böker, H, Hättenschwiler, J, et al. The association of interoceptive awareness and alexithymia with neurotransmitter concentrations in insula and anterior cingulate. Social Cognitive and Affective Neuroscience. 2013; 9(6): 857863.Google Scholar
Deng, Y, Ma, X, Tang, Q. Brain response during visual emotional processing: An fMRI study of alexithymia. Psychiatry Research: Neuroimaging. 2013; 213(3): 225229.Google Scholar
Goerlich-Dobre, KS, Bruce, L, Martens, S, Aleman, A, Hooker, CI. Distinct associations of insula and cingulate volume with the cognitive and affective dimensions of alexithymia. Neuropsychologia. 2014; 53: 284292.Google Scholar
Hogeveen, J, Bird, G, Chau, A, Krueger, F, Grafman, J. Acquired alexithymia following damage to the anterior insula. Neuropsychologia. 2016; 82: 142148.Google Scholar
Zotev, V, Krueger, F, Phillips, R, et al. Self-regulation of amygdala activation using real-time fMRI neurofeedback. PloS One. 2011; 6(9): e24522.Google Scholar
MacDuffie, KE, MacInnes, J, Dickerson, KC, et al. Single session real-time fMRI neurofeedback has a lasting impact on cognitive behavioral therapy strategies. NeuroImage: Clinical. 2018; 19: 868875.Google Scholar
Muris, P. Relationships between self-efficacy and symptoms of anxiety disorders and depression in a normal adolescent sample. Personality and Individual Differences. 2002; 32(2): 337348.CrossRefGoogle Scholar
Kavanagh, DJ, Wilson, PH. Prediction of outcome with group cognitive therapy for depression. Behaviour Research and Therapy. 1989; 27(4): 333343.Google Scholar
Maddux, JE, Meier, LJ. Self-Efficacy and Depression. Self-Efficacy, Adaptation, and Adjustment. Springer; 1995. 143169.CrossRefGoogle Scholar
Bartra, O, McGuire, JT, Kable, JW. The valuation system: A coordinate-based meta-analysis of BOLD fMRI experiments examining neural correlates of subjective value. Neuroimage. 2013; 76: 412427.Google Scholar
Balleine, BW, Delgado, MR, Hikosaka, O. The role of the dorsal striatum in reward and decision-making. Journal of Neuroscience. 2007; 27(31): 81618165.Google Scholar
Young, KD, Siegle, GJ, Misaki, M, et al. Altered task-based and resting-state amygdala functional connectivity following real-time fMRI amygdala neurofeedback training in major depressive disorder. NeuroImage: Clinical. 2018; 17: 691703.Google Scholar
Sheline, YI, Barch, DM, Price, JL, et al. The default mode network and self-referential processes in depression. Proceedings of the National Academy of Sciences. 2009; 106(6): 19421947.Google Scholar
Zhu, X, Wang, X, Xiao, J, et al. Evidence of a dissociation pattern in resting-state default mode network connectivity in first-episode, treatment-naive major depression patients. Biological Psychiatry. 2012; 71(7): 611617.Google Scholar
Hamilton, JP, Furman, DJ, Chang, C, et al. Default-mode and task-positive network activity in major depressive disorder: Implications for adaptive and maladaptive rumination. Biological Psychiatry. 2011; 70(4): 327333.Google Scholar
Marchetti, I, Koster, EH, Sonuga-Barke, EJ, De Raedt, R. The default mode network and recurrent depression: A neurobiological model of cognitive risk factors. Neuropsychology Review. 2012; 22(3): 229251.Google Scholar
Belleau, EL, Taubitz, LE, Larson, CL. Imbalance of default mode and regulatory networks during externally focused processing in depression. Social Cognitive and Affective Neuroscience. 2014; 10(5): 744751.Google Scholar
Burkhouse, KL, Jacobs, RH, Peters, AT, et al. Neural correlates of rumination in adolescents with remitted major depressive disorder and healthy controls. Cognitive, Affective, & Behavioral Neuroscience. 2017; 17(2): 394405.Google Scholar
Mandell, D, Siegle, GJ, Shutt, L, Feldmiller, J, Thase, ME. Neural substrates of trait ruminations in depression. Journal of Abnormal Psychology. 2014; 123(1): 35.CrossRefGoogle ScholarPubMed
Siegle, GJ, Steinhauer, SR, Thase, ME, Stenger, VA, Carter, CS. Can’t shake that feeling: event-related fMRI assessment of sustained amygdala activity in response to emotional information in depressed individuals. Biological Psychiatry. 2002; 51(9): 693707.CrossRefGoogle ScholarPubMed
Siegle, GJ, Carter, CS, Thase, ME. Use of fMRI to predict recovery from unipolar depression with cognitive behavior therapy. American Journal of Psychiatry. 2006; 163(4): 735738.Google Scholar
Linden, DE, Habes, I, Johnston, SJ, et al. Real-time self-regulation of emotion networks in patients with depression. PloS One. 2012; 7(6): e38115.Google Scholar
Yuan, H, Young, KD, Phillips, R, et al. Resting-state functional connectivity modulation and sustained changes after real-time functional magnetic resonance imaging neurofeedback training in depression. Brain Connectivity. 2014; 4(9): 690701.Google Scholar
Hamilton, JP, Glover, GH, Bagarinao, E, et al. Effects of salience-network-node neurofeedback training on affective biases in major depressive disorder. Psychiatry Research: Neuroimaging. 2016; 249: 9196.Google Scholar
Teasdale, JD, Green, HA. Ruminative self-focus and autobiographical memory. Personality and Individual Differences. 2004; 36(8): 19331943.Google Scholar
Sutherland, K, Bryant, RA. Rumination and overgeneral autobiographical memory. Behaviour Research and Therapy. 2007; 45(10): 24072416.Google Scholar
Ramot, M, Grossman, S, Friedman, D, Malach, R. Covert neurofeedback without awareness shapes cortical network spontaneous connectivity. Proceedings of the National Academy of Sciences. 2016; 113(17): E2413–E20.Google Scholar
Thibault, RT, MacPherson, A, Lifshitz, M, Roth, RR, Raz, A. Neurofeedback with fMRI: A critical systematic review. Neuroimage. 2018; 172: 786807.Google Scholar
Hamann, S, Canli, T. Individual differences in emotion processing. Current Opinion in Neurobiology. 2004; 14(2): 233238.Google Scholar
Berlim, MT, Van den Eynde, F, Daskalakis, ZJ. Clinically meaningful efficacy and acceptability of low-frequency repetitive transcranial magnetic stimulation (rTMS) for treating primary major depression: a meta-analysis of randomized, double-blind and sham-controlled trials. Neuropsychopharmacology. 2013; 38(4): 543.CrossRefGoogle ScholarPubMed
Berlim, M, Van den Eynde, F, Tovar-Perdomo, S, Daskalakis, Z. Response, remission and drop-out rates following high-frequency repetitive transcranial magnetic stimulation (rTMS) for treating major depression: A systematic review and meta-analysis of randomized, double-blind and sham-controlled trials. Psychological Medicine. 2014; 44(2): 225239.Google Scholar
Young, KD, Zotev, V, Phillips, R, Misaki, M, Yuan, H, Drevets, WC, et al. Real-time fMRI neurofeedback training of amygdala activity in patients with major depressive disorder. PloS One. 2014; 9(2):e88785.Google Scholar
Yang, Y, Zhong, N, Imamura, K, et al. Task and resting-state fMRI reveal altered salience responses to positive stimuli in patients with major depressive disorder. PLOS ONE. 2016; 11(5): e0155092.Google Scholar
Sheline, YI, Barch, DM, Donnelly, JM, et al. Increased amygdala response to masked emotional faces in depressed subjects resolves with antidepressant treatment: An fMRI study. Biological Psychiatry. 2001; 50(9): 651658.Google Scholar
Drevets, WC, Price, JL, Bardgett, ME, et al. Glucose metabolism in the amygdala in depression: Relationship to diagnostic subtype and plasma cortisol levels. Pharmacology Biochemistry and Behavior. 2002; 71(3): 431447.Google Scholar
Drevets, WC. Neuroimaging abnormalities in the amygdala in mood disorders. Annals of the New York Academy of Sciences. 2003; 985(1): 420444.Google Scholar
Victor, TA, Furey, ML, Fromm, SJ, Öhman, A, Drevets, WC. Relationship between amygdala responses to masked faces and mood state and treatment in major depressive disorder. Archives of General Psychiatry. 2010; 67(11): 11281138.Google Scholar
Suslow, T, Konrad, C, Kugel, H, et al. Automatic mood-congruent amygdala responses to masked facial expressions in major depression. Biological Psychiatry. 2010; 67(2): 155160.Google Scholar
Seeley, WW, Menon, V, Schatzberg, AF, Keller, J, Glover, GH, Kenna, H, et al. Dissociable intrinsic connectivity networks for salience processing and executive control. Journal of Neuroscience. 2007; 27(9): 2349–56.Google Scholar
Jacobs, R, Barba, A, Gowins, J, et al. Decoupling of the amygdala to other salience network regions in adolescent-onset recurrent major depressive disorder. Psychological Medicine. 2016; 46(5): 10551067.Google Scholar
Packard, MG, Cahill, L. Affective modulation of multiple memory systems. Current Opinion in Neurobiology. 2001; 11(6): 752756.Google Scholar
Burianova, H, McIntosh, AR, Grady, CL. A common functional brain network for autobiographical, episodic, and semantic memory retrieval. Neuroimage. 2010; 49(1): 865874.Google Scholar
Noulhiane, M, Piolino, P, Hasboun, D, et al. Autobiographical memory after temporal lobe resection: neuropsychological and MRI volumetric findings. Brain. 2007; 130(12): 31843199.Google Scholar
Aggleton, JP, Brown, MW. Episodic memory, amnesia, and the hippocampal–anterior thalamic axis. Behavioral and Brain Sciences. 1999; 22(3): 425444.Google Scholar
Young, KD. Effects of amygdala neurofeedback on depressive symptoms. Identification No. NCT02709161. Retrieved from https://clinicaltrials.gov/ct2/show/NCT02709161. 2016.Google Scholar
Young, KD. Neurofeedback for treatment resistant depression. Identification No. NCT03428828. Retrieved from https://clinicaltrials.gov/ct2/show/NCT03428828. 2018.Google Scholar
Engen, HG, Bernhardt, BC, Skottnik, L, Ricard, M, Singer, T. Structural changes in socio-affective networks: Multi-modal MRI findings in long-term meditation practitioners. Neuropsychologia. 2018; 116: 2633.Google Scholar
Young, KD, Misaki, M, Harmer, CJ, et al. Real-time functional magnetic resonance imaging amygdala neurofeedback changes positive information processing in major depressive disorder. Biological Psychiatry. 2017; 82(8): 578586.Google Scholar
Mathiak, K. Symptom based treatment affects brain plasticity – cognitive training in patients with affective symptoms (APIC-II). Identification No. NCT03183947. Retrieved from https://clinicaltrials.gov/ct2/show/NCT03183947. 2017.Google Scholar
Hamilton, JP, Etkin, A, Furman, DJ, et al. Functional neuroimaging of major depressive disorder: A meta-analysis and new integration of baseline activation and neural response data. American Journal of Psychiatry. 2012; 169(7): 693703.Google Scholar
Caria, A, Sitaram, R, Veit, R, Begliomini, C, Birbaumer, N. Volitional control of anterior insula activity modulates the response to aversive stimuli. A real-time functional magnetic resonance imaging study. Biological Psychiatry. 2010; 68(5): 425432.Google Scholar
Herwig, U, Lutz, J, Scherpiet, S, et al. Training emotion regulation through real-time fMRI neurofeedback of amygdala activity. NeuroImage. 2019; 184: 687696.Google Scholar
Perlman, G, Simmons, AN, Wu, J, et al. Amygdala response and functional connectivity during emotion regulation: a study of 14 depressed adolescents. Journal of Affective Disorders. 2012; 139(1): 7584.Google Scholar
Carballedo, A, Scheuerecker, J, Meisenzahl, E, et al. Functional connectivity of emotional processing in depression. Journal of Affective Disorders. 2011; 134(1–3): 272279.Google Scholar
de Almeida, JRC, Versace, A, Mechelli, A, et al. Abnormal amygdala-prefrontal effective connectivity to happy faces differentiates bipolar from major depression. Biological Psychiatry. 2009; 66(5): 451459.Google Scholar
Zahn, R, Weingartner, J, Basilio, R, et al. 30 Blame Rebalance fMRI Feedback Proof-of-Concept Trial in Major Depressive Disorder. BMJ Publishing Group Ltd; 2017.Google Scholar
Greicius, MD, Flores, BH, Menon, V, et al. Resting-state functional connectivity in major depression: Abnormally increased contributions from subgenual cingulate cortex and thalamus. Biological Psychiatry. 2007; 62(5): 429437.CrossRefGoogle ScholarPubMed
Sheline, YI, Price, JL, Yan, Z, Mintun, MA. Resting-state functional MRI in depression unmasks increased connectivity between networks via the dorsal nexus. Proceedings of the National Academy of Sciences. 2010; 107(24): 1102011025.Google Scholar
Veer, IM, Beckmann, C, Van Tol, M-J, et al. Whole brain resting-state analysis reveals decreased functional connectivity in major depression. Frontiers in Systems Neuroscience. 2010; 4: 41.Google Scholar
Drysdale, AT, Grosenick, L, Downar, J, et al. Resting-state connectivity biomarkers define neurophysiological subtypes of depression. Nature Medicine. 2017; 23(1): 28.Google Scholar
Dinga, R, Schmaal, L, Penninx, B, et al. Evaluating the evidence for biotypes of depression: Methodological replication and extension of. NeuroImage: Clinical. 2019; 22: 101796.CrossRefGoogle ScholarPubMed
Yamada, T, Hashimoto, R-i, Yahata, N, et al. Resting-state functional connectivity-based biomarkers and functional MRI-based neurofeedback for psychiatric disorders: a challenge for developing theranostic biomarkers. International Journal of Neuropsychopharmacology. 2017; 20(10): 769781.Google Scholar
Moll, J, Weingartner, JH, Bado, P, et al. Voluntary enhancement of neural signatures of affiliative emotion using fMRI neurofeedback. PloS One. 2014; 9(5): e97343.Google Scholar
Sokhadze, EM, El-Baz, AS, Tasman, A, et al. Neuromodulation integrating rTMS and neurofeedback for the treatment of autism spectrum disorder: an exploratory study. Applied Psychophysiology and Biofeedback. 2014; 39(3–4): 237257.Google Scholar
Koganemaru, S, Mikami, Y, Maezawa, H, et al. Neurofeedback control of the human GABAergic system using non-invasive brain stimulation. Neuroscience. 2018; 380: 3848.Google Scholar
Siepmann, M, Aykac, V, Unterdörfer, J, Petrowski, K, Mueck-Weymann, M. A pilot study on the effects of heart rate variability biofeedback in patients with depression and in healthy subjects. Applied Psychophysiology and Biofeedback. 2008; 33(4): 195201.CrossRefGoogle ScholarPubMed
Karavidas, MK, Lehrer, PM, Vaschillo, E, et al. Preliminary results of an open label study of heart rate variability biofeedback for the treatment of major depression. Applied Psychophysiology and Biofeedback. 2007; 32(1): 1930.CrossRefGoogle ScholarPubMed
Gevirtz, R. The promise of heart rate variability biofeedback: Evidence-based applications. Biofeedback. 2013; 41(3): 110120.Google Scholar
Tang, Y, Posner, MI, Rothbart, MK. Meditation improves self‐regulation over the life span. Annals of the New York Academy of Sciences. 2014; 1307(1): 104111.Google Scholar
Tang, Y, Ma, Y, Wang, J, et al. Short-term meditation training improves attention and self-regulation. Proceedings of the National Academy of Sciences. 2007; 104(43): 1715217156.Google Scholar
Baird, B, Mrazek, MD, Phillips, DT, Schooler, JW. Domain-specific enhancement of metacognitive ability following meditation training. Journal of Experimental Psychology: General. 2014; 143(5): 1972.Google Scholar
Hofmann, SG, Sawyer, AT, Witt, AA, Oh, D. The effect of mindfulness-based therapy on anxiety and depression: A meta-analytic review. Journal of Consulting and Clinical Psychology. 2010; 78(2): 169.Google Scholar
Peciña, M. Study of neural responses induced by antidepressant effects (SONRISA). Identification No. NCT02674529. Retrieved from https://clinicaltrials.gov/ct2/show/NCT02674529. 2016.Google Scholar
Scharnowski, F. Neural correlates of neurofeedback training. Identification No. NCT03165578. Retrieved from https://clinicaltrials.gov/ct2/show/NCT03165578. 2017.Google Scholar
Sorger, B, Scharnowski, F, Linden, DE, Hampson, M, Young, KD. Control freaks: Towards optimal selection of control conditions for fMRI neurofeedback studies. NeuroImage. 2019; 186: 256265.Google Scholar
Randell, E, McNamara, R, Subramanian, L, Hood, K, Linden, D. Current Practices in Clinical Neurofeedback with Functional MRI-Analysis of a Survey Using the TIDieR Checklist. Eur Psychiatry; 2018.Google Scholar
Grossberg, , S, On the dynamics of operant conditioning. Journal of Theoretical Biology. 1971 November; 33(2): 225255.Google Scholar
Staddon, JER, Cerutti, DT. Operant Conditioning. Annu Rev Psychol. 2003; 54: 115144.Google Scholar

References

Jobsis, FF. Noninvasive, infrared monitoring of cerebral and myocardial oxygen sufficiency and circulatory parameters. Science. 1977; 198(4323): 12641267.Google Scholar
Villringer, A, Planck, J, Hock, C, Schleinkofer, L, Dirnagl, U. Near infrared spectroscopy (NIRS): A new tool to study hemodynamic changes during activation of brain function in human adults. Neurosci Lett. 1993; 154(1–2): 101104.Google Scholar
Gratton, G, Maier, JS, Fabiani, M, Mantulin, WW, Gratton, E. Feasibility of intracranial near-infrared optical scanning. Psychophysiology. 1994; 31(2): 211215.CrossRefGoogle ScholarPubMed
Maki, A, Yamashita, Y, Ito, Y, et al. Spatial and temporal analysis of human motor activity using noninvasive NIR topography. Med Phys. 1995; 22(12): 19972005.CrossRefGoogle ScholarPubMed
Villringer, A, Chance, B. Non-invasive optical spectroscopy and imaging of human brain function. Trends Neurosci. 1997; 20(10): 435442.Google Scholar
Obrig, H. et al. Near-infrared spectroscopy: Does it function in functional activation studies of the adult brain? Int J Psychophysiol. 2000; 35(2–3): 125142.CrossRefGoogle ScholarPubMed
Yamashita, Y, Maki, A, Koizumi, H. Wavelength dependence of the precision of noninvasive optical measurement of oxy-, deoxy-, and total-hemoglobin concentration. Med Phys. 2001; 28(6): 11081114.Google Scholar
Ferrari, M, Quaresima, V. A brief review on the history of human functional near-infrared spectroscopy (fNIRS) development and fields of application. Neuroimage. 2012; 63(2): 921935.Google Scholar
Strangman, G, Boas, DA, Sutton, JP. Non-invasive neuroimaging using near-infrared light. Biol Psychiatry. 2002; 52(7): 679693.Google Scholar
Elwell, CE. et al. Measurement of adult cerebral haemodynamics using near infrared spectroscopy. Acta Neurochir Suppl (Wien). 1993; 59: 7480.Google ScholarPubMed
Takahashi, T, Takikawa, Y, Kawagoe, R, et al. Influence of skin blood flow on near-infrared spectroscopy signals measured on the forehead during a verbal fluency task. Neuroimage. 2011; 57(3): 9911002.Google Scholar
Kohri, S, Hoshi, Y, Tamura, M, et al. Quantitative evaluation of the relative contribution ratio of cerebral tissue to near-infrared signals in the adult human head: A preliminary study. Physiol Meas. 2002; 23(2): 301312.Google Scholar
Fox, PT, Raichle, ME. Focal physiological uncoupling of cerebral blood flow and oxidative metabolism during somatosensory stimulation in human subjects. Proc Natl Acad Sci U S A. 1986; 83(4): 11401144.CrossRefGoogle ScholarPubMed
Hock, C. et al. Decrease in parietal cerebral hemoglobin oxygenation during performance of a verbal fluency task in patients with Alzheimer’s disease monitored by means of near-infrared spectroscopy (NIRS)–correlation with simultaneous rCBF-PET measurements. Brain Res. 1997; 755(2): 293303.Google Scholar
Sato, H. et al. A NIRS-fMRI investigation of prefrontal cortex activity during a working memory task. Neuroimage. 2013; 83: 158173.Google Scholar
Cui, X, Bray, S, Bryant, DM, Glover, GH, Reiss, AL. A quantitative comparison of NIRS and fMRI across multiple cognitive tasks. Neuroimage. 2011; 54(4): 28082821.Google Scholar
Strangman, G, Culver, JP, Thompson, JH, Boas, DA. A quantitative comparison of simultaneous BOLD fMRI and NIRS recordings during functional brain activation. Neuroimage. 2002; 17(2): 719731.Google Scholar
Moriguchi, Y. et al. Validation of brain-derived signals in near-infrared spectroscopy through multivoxel analysis of concurrent functional magnetic resonance imaging. Hum Brain Mapp. 2017; 38(10): 52745291.Google Scholar
Tsuzuki, D, Jurcak, V, Singh, AK, et al. Virtual spatial registration of stand-alone fNIRS data to MNI space. Neuroimage. 2007; 34(4): 15061518.Google Scholar
Okada, F, Takahashi, N, Tokumitsu, Y. Dominance of the ‘nondominant’ hemisphere in depression. J Affect Disord. 1996; 37(1): 1321.CrossRefGoogle ScholarPubMed
Cyranoski, D. Neuroscience: Thought experiment. Nature. 2011; 469(7329): 148149.Google Scholar
Zhang, H. et al. Near-infrared spectroscopy for examination of prefrontal activation during cognitive tasks in patients with major depressive disorder: A meta-analysis of observational studies. Psychiatry Clin Neurosci. 2015; 69(1): 2233.Google Scholar
Matsuo, K, Kato, T, Fukuda, M, Kato, N. Alteration of hemoglobin oxygenation in the frontal region in elderly depressed patients as measured by near-infrared spectroscopy. J Neuropsychiatry Clin Neurosci. 2000; 12(4): 465471.Google Scholar
Suto, T, Fukuda, M, Ito, M, Uehara, T, Mikuni, M. Multichannel near-infrared spectroscopy in depression and schizophrenia: cognitive brain activation study. Biol Psychiatry. 2004; 55(5): 501511.Google Scholar
Hirano, J. et al. Frontal and temporal cortical functional recovery after electroconvulsive therapy for depression: A longitudinal functional near-infrared spectroscopy study. J Psychiatr Res. 2017; 91: 2635.CrossRefGoogle ScholarPubMed
Koseki, S. et al. The relationship between positive and negative automatic thought and activity in the prefrontal and temporal cortices: A multi-channel near-infrared spectroscopy (NIRS) study. J Affect Disord. 2013; 151(1): 352359.CrossRefGoogle ScholarPubMed
Noda, T. et al. Frontal and right temporal activations correlate negatively with depression severity during verbal fluency task: A multi-channel near-infrared spectroscopy study. J Psychiatr Res. 2012; 46(7): 905912.CrossRefGoogle ScholarPubMed
Pu, S. et al. The relationship between the prefrontal activation during a verbal fluency task and stress-coping style in major depressive disorder: A near-infrared spectroscopy study. J Psychiatr Res. 2012; 46(11): 14271434.Google Scholar
Pu, S. et al. Reduced frontopolar activation during verbal fluency task associated with poor social functioning in late-onset major depression: Multi-channel near-infrared spectroscopy study. Psychiatry Clin Neurosci. 2008; 62(6): 728737.Google Scholar
Tsujii, N. et al. Relationship between prefrontal hemodynamic responses and quality of life differs between melancholia and non-melancholic depression. Psychiatry Res. 2016; 253(30): 2635.Google Scholar
Usami, M, Iwadare, Y, Kodaira, M, Watanabe, K, Saito, K. Near infrared spectroscopy study of the frontopolar hemodynamic response and depressive mood in children with major depressive disorder: A pilot study. PLoS One. 2014; 9(1): e86290.Google Scholar
Akashi, H, Tsujii, N, Mikawa, W, et al. Prefrontal cortex activation is associated with a discrepancy between self- and observer-rated depression severities of major depressive disorder: A multichannel near-infrared spectroscopy study. J Affect Disord. 2015; 174(15): 165172.Google Scholar
Tsujii, N. et al. Right temporal activation differs between melancholia and nonmelancholic depression: A multichannel near-infrared spectroscopy study. J Psychiatr Res. 2014; 55: 17.Google Scholar
Pu, S. et al. Suicidal ideation is associated with reduced prefrontal activation during a verbal fluency task in patients with major depressive disorder. J Affect Disord. 2015; 18: 917.Google Scholar
Tomioka, H. et al. A longitudinal functional neuroimaging study in medication-naive depression after antidepressant treatment. PLoS One. 2015; 10(3): e0120828.Google Scholar
Masuda, K. et al. Different functioning of prefrontal cortex predicts treatment response after a selective serotonin reuptake inhibitor treatment in patients with major depression. J Affect Disord. 2017; 214: 4452.Google Scholar
Nishida, M, Kikuchi, S, Matsumoto, K, et al. Sleep complaints are associated with reduced left prefrontal activation during a verbal fluency task in patients with major depression: A multi-channel near-infrared spectroscopy study. J Affect Disord. 2017; 207: 102109.Google Scholar
Takamiya, A. et al. High-dose antidepressants affect near-infrared spectroscopy signals: A retrospective study. Neuroimage Clin. 2017; 14: 648655.Google Scholar
Tsujii, N. et al. Reduced left precentral regional responses in patients with major depressive disorder and history of suicide attempts. PLoS One. 2017; 12(4): e0175249.CrossRefGoogle ScholarPubMed
Schlosser, R. et al. Functional magnetic resonance imaging of human brain activity in a verbal fluency task. J Neurol Neurosurg Psychiatry. 1998; 64(4): 492498.Google Scholar
Cuenod, CA, Bookheimer, SY, Hertz-Pannier, L, et al. Functional MRI during word generation, using conventional equipment: A potential tool for language localization in the clinical environment,. Neurology. 1995; 45(10): 18211827.Google Scholar
Strauss, E, Sherman, EMS, Spreen, O, Spreen, O. A Compendium of Neuropsychological Tests : Administration, Norms, and Commentary. Oxford; New York: Oxford University Press, 2006.Google Scholar
Ma, XY. et al. Near-infrared spectroscopy reveals abnormal hemodynamics in the left dorsolateral prefrontal cortex of menopausal depression patients. Dis Markers. 2017; (2017): 1695930. DOI: 10.1155/2017/1695930Google Scholar
Liu, X. et al. Relationship between the prefrontal function and the severity of the emotional symptoms during a verbal fluency task in patients with major depressive disorder: A multi-channel NIRS study. Prog Neuropsychopharmacol Biol Psychiatry. 2014; 54: 114121.Google Scholar
Pu, S. et al. Reduced prefrontal cortex activation during the working memory task associated with poor social functioning in late-onset depression: Multi-channel near-infrared spectroscopy study. Psychiatry Res. 2012; 203(2–3): 222228.Google Scholar
Eschweiler, GW. et al. Left prefrontal activation predicts therapeutic effects of repetitive transcranial magnetic stimulation (rTMS) in major depression. Psychiatry Res. 2000; 99(3): 161172.Google Scholar
Pu, S. et al. A multi-channel near-infrared spectroscopy study of prefrontal cortex activation during working memory task in major depressive disorder. Neurosci Res. 2011; 70(1): 9197.Google Scholar
Onishi, Y, Kikuchi, S, Watanabe, E, Kato, S. Alterations in prefrontal cortical activity in the course of treatment for late-life depression as assessed on near-infrared spectroscopy. Psychiatry Clin Neurosci. 2008; 62(2): 177184.Google Scholar
Kikuchi, S. et al. Prefrontal cerebral activity during a simple “rock, paper, scissors” task measured by the noninvasive near-infrared spectroscopy method. Psychiatry Res. 2007; 156(3): 199208.Google Scholar
Matsuo, K, Kato, N, Kato, T. Decreased cerebral haemodynamic response to cognitive and physiological tasks in mood disorders as shown by near-infrared spectroscopy. Psychol Med. 2002; 32(6): 10291037.Google Scholar
Matsuo, K, Onodera, Y, Hamamoto, T, et al. Hypofrontality and microvascular dysregulation in remitted late-onset depression assessed by functional near-infrared spectroscopy. Neuroimage. 2005; 26(1): 234242.Google Scholar
Ono, Y. et al. Prefrontal oxygenation during verbal fluency and cognitive function in adolescents with bipolar disorder type II. Asian J Psychiatr. 2017; 25: 147153.Google Scholar
Mikawa, W, Tsujii, N, Akashi, H, et al. Left temporal activation associated with depression severity during a verbal fluency task in patients with bipolar disorder: A multichannel near-infrared spectroscopy study. J Affect Disord. 2015; 173: 193200.Google Scholar
Nishimura, Y. et al. Social function and frontopolar activation during a cognitive task in patients with bipolar disorder. Neuropsychobiology. 2015; 72(2): 8190.CrossRefGoogle ScholarPubMed
Nishimura, Y, Takahashi, K, Ohtani, T, et al. Dorsolateral prefrontal hemodynamic responses during a verbal fluency task in hypomanic bipolar disorder. Bipolar Disord. 2015; 17(2): 172183.Google Scholar
Ono, Y. et al. Reduced prefrontal activation during performance of the Iowa gambling task in patients with bipolar disorder. Psychiatry Res. 2015; 233(1): 18.Google Scholar
Matsuo, K. et al. A near-infrared spectroscopy study of prefrontal cortex activation during a verbal fluency task and carbon dioxide inhalation in individuals with bipolar disorder. Bipolar Disord. 2007; 9(8): 876883.Google Scholar
Matsuo, K, Watanabe, A, Onodera, Y, Kato, N, Kato, T. Prefrontal hemodynamic response to verbal-fluency task and hyperventilation in bipolar disorder measured by multi-channel near-infrared spectroscopy. J Affect Disord. 2004; 82(1): 8592.Google Scholar
Kubota, Y. et al. Altered prefrontal lobe oxygenation in bipolar disorder: A study by near-infrared spectroscopy. Psychol Med. 2009; 39(8): 12651275.CrossRefGoogle ScholarPubMed
Ohi, K. et al. Impact of familial loading on prefrontal activation in major psychiatric disorders: A near-infrared spectroscopy (NIRS) study. Sci Rep. 2017; 7: 44268.Google Scholar
Ohta, H. et al. Hypofrontality in panic disorder and major depressive disorder assessed by multi-channel near-infrared spectroscopy. Depress Anxiety. 2008; 25(12): 10531059.Google Scholar
Kameyama, M. et al. Frontal lobe function in bipolar disorder: A multichannel near-infrared spectroscopy study. Neuroimage. 2006; 29(1): 172184.Google Scholar
Takizawa, R. et al. Neuroimaging-aided differential diagnosis of the depressive state. Neuroimage. 2014; 85(Pt 1): 498507.Google Scholar
Kinou, M. et al. Differential spatiotemporal characteristics of the prefrontal hemodynamic response and their association with functional impairment in schizophrenia and major depression. Schizophr Res. 2013; 150(2–3): 459467.Google Scholar
Ohtani, T, Nishimura, Y, Takahashi, K, et al. Association between longitudinal changes in prefrontal hemodynamic responses and social adaptation in patients with bipolar disorder and major depressive disorder. J Affect Disord. 2015; 176: 7886.Google Scholar
Zhu, Y. et al. Prefrontal activation during a working memory task differs between patients with unipolar and bipolar depression: A preliminary exploratory study. J Affect Disord. 2018 January 1; 225: 6470.Google Scholar
Matsubara, T. et al. Prefrontal activation in response to emotional words in patients with bipolar disorder and major depressive disorder. Neuroimage. 2014; 85(Pt 1): 489497.CrossRefGoogle ScholarPubMed
Uceyler, N, Zeller, J, Kewenig, S, et al. Increased cortical activation upon painful stimulation in fibromyalgia syndrome. BMC Neurol. 2015; 15: 210.Google Scholar
Takei, Y. et al. Near-infrared spectroscopic study of frontopolar activation during face-to-face conversation in major depressive disorder and bipolar disorder. J Psychiatr Res. 2014; 57: 7483.Google Scholar

References

Woodman, GF. A brief introduction to the use of event-related potentials in studies of perception and attention. Atten Percept Psychophys [Internet]. 2010 November; 72(8): 20312046. Available from: www.ncbi.nlm.nih.gov/pubmed/21097848CrossRefGoogle Scholar
Vandoolaeghe, E, van Hunsel, F, Nuyten, D, Maes, M. Auditory event related potentials in major depression: prolonged P300 latency and increased P200 amplitude. J Affect Disord. 1998 March; 48(2–3): 105113.Google Scholar
Pfefferbaum, A, Wenegrat, BG, Ford, JM, Roth, WT, Kopell, BS. Clinical application of the P3 component of event-related potentials. II. Dementia, depression and schizophrenia. Electroencephalogr Clin Neurophysiol. 1984 April; 59(2): 104124.Google Scholar
Diner, BC, Holcomb, PJ, Dykman, RA. P300 in major depressive disorder. Psychiatry Res. 1985 July; 15(3): 175184.Google Scholar
Bange, F, Bathien, N. Visual cognitive dysfunction in depression: An event-related potential study. Electroencephalogr Clin Neurophysiol. 1998 September; 108(5): 472481.CrossRefGoogle ScholarPubMed
Schlegel, S, Nieber, D, Herrmann, C, Bakauski, E. Latencies of the P300 component of the auditory event-related potential in depression are related to the Bech-Rafaelsen melancholia scale but not to the Hamilton Rating Scale for Depression. Acta Psychiatr Scand. 1991 June; 83(6): 438440.Google Scholar
Pierson, A, Ragot, R, Van Hooff, J, et al. Heterogeneity of information-processing alterations according to dimensions of depression: an event-related potentials study. Biol Psychiatry. 1996; 40(2): 98115.Google Scholar
Bruder, GE, Kroppmann, CJ, Kayser, J, et al. Reduced brain responses to novel sounds in depression: P3 findings in a novelty oddball task. Psychiatry Res. 2009; 170(2–3): 218223.Google Scholar
Friedman, D, Simpson, G, Hamberger, M. Age‐related changes in scalp topography to novel and target stimuli. Psychophysiology. 1993; 30(4): 383396.CrossRefGoogle ScholarPubMed
Tenke, CE, Kayser, J, Stewart, JW, Bruder, GE. Novelty P3 reductions in depression: characterization using principal components analysis (PCA) of current source density (CSD) waveforms. Psychophysiology. 2010; 47(1): 133146.CrossRefGoogle ScholarPubMed
Fu, L, Xiang, D, Subodh, D, et al. Auditory P300 study in patients with convalescent bipolar depression and bipolar depression. Neuroreport. 2018 August; 29(11): 968973.Google Scholar
Bersani, FS, Minichino, A, Fattapposta, F, et al. P300 component in euthymic patients with bipolar disorder type I, bipolar disorder type II and healthy controls: A preliminary event-related potential study. Neuroreport. 2015 March; 26(4): 206210.Google Scholar
Bruder, GE, Kayser, J, Tenke, CCE. Event-related brain potentials in depression: clinical, cognitive and neurophysiologic implications. Oxford Handb event-related potential components [Internet]. 2012; 2012: 563592. Available from: http://psychophysiology.cpmc.columbia.edu/pdf/bruder2009a.pdfGoogle Scholar
Karaaslan, F, Gonul, AS, Oguz, A, Erdinc, E, Esel, E. P300 changes in major depressive disorders with and without psychotic features. J Affect Disord. 2003; 73(3): 283287.Google Scholar
Li, Y, Hu, Y, Liu, T, Wu, D. Dipole source analysis of auditory P300 response in depressive and anxiety disorders. Cogn Neurodyn. 2011; 5(2): 221229.Google Scholar
Li, Y, Wang, W, Liu, T, et al. Source analysis of P3a and P3b components to investigate interaction of depression and anxiety in attentional systems. Sci Rep. 2015; 5: 17138.CrossRefGoogle ScholarPubMed
Hetzel, G, Moeller, O, Evers, S, et al. The astroglial protein S100B and visually evoked event-related potentials before and after antidepressant treatment. Psychopharmacology (Berl). 2005; 178(2–3): 161166.Google Scholar
Hansenne, M, Ansseau, M. P300 event-related potential and serotonin-1A activity in depression. Eur psychiatry. 1999; 14(3): 143147.Google Scholar
Jaworska, N, Protzner, A. Electrocortical features of depression and their clinical utility in assessing antidepressant treatment outcome. Can J Psychiatry. 2013; 58(9): 509514.Google Scholar
Arns, M, Drinkenburg, WH, Fitzgerald, PB, Kenemans, JL. Neurophysiological predictors of non-response to rTMS in depression. Brain Stimul [Internet]. 2012; 5(4): 569576. Available from: http://dx.doi.org/10.1016/j.brs.2011.12.003Google Scholar
Tripathi, SM, Mishra, N, Tripathi, RK, Gurnani, KC. P300 latency as an indicator of severity in major depressive disorder. Ind Psychiatry J. 2015; 24(2): 163167.CrossRefGoogle ScholarPubMed
Scheffers, MK, Coles, MGH. Performance monitoring in a confusing world: Error-related brain activity, judgments of response accuracy, and types of errors. Vol. 26, Journal of Experimental Psychology: Human Perception and Performance. US: American Psychological Association; 2000. 141151.Google Scholar
Olvet, DM, Hajcak, G. The error-related negativity (ERN) and psychopathology: Toward an Endophenotype. Clin Psychol Rev [Internet]. 2008 December 9; 28(8): 13431354. Available from: www.ncbi.nlm.nih.gov/pmc/articles/PMC2615243/Google Scholar
Schoenberg, PLA. The error processing system in major depressive disorder: Cortical phenotypal marker hypothesis. Biol Psychol. 2014 May; 99: 100114.Google Scholar
Ruchsow, M, Herrnberger, B, Wiesend, C, et al. The effect of erroneous responses on response monitoring in patients with major depressive disorder: A study with event-related potentials. Psychophysiology. 2004 November; 41(6): 833840.Google Scholar
Schrijvers, D, Hulstijn, W, Sabbe, BGC. Psychomotor symptoms in depression: A diagnostic, pathophysiological and therapeutic tool. J Affect Disord. 2008 July; 109(1–2): 120.Google Scholar
Schrijvers, D, de Bruijn, ERA, Maas, Y, et al. Action monitoring in major depressive disorder with psychomotor retardation. Cortex. 2008 May; 44(5): 569579.Google Scholar
Schrijvers, D, De Bruijn, ERA, Maas, YJ, et al. Action monitoring and depressive symptom reduction in major depressive disorder. Int J Psychophysiol. 2009 March; 71(3): 218224.CrossRefGoogle ScholarPubMed
Weinberg, A, Liu, H, Shankman, SA. Blunted neural response to errors as a trait marker of melancholic depression. Biol Psychol. 2016 January; 113: 100107.Google Scholar
Chiu, PH, Deldin, PJ. Neural evidence for enhanced error detection in major depressive disorder. Am J Psychiatry. 2007; 164(4): 608616.Google Scholar
Gorka, SM, Lieberman, L, Shankman, SA, Phan, KL. Startle potentiation to uncertain threat as a psychophysiological indicator of fear-based psychopathology: An examination across multiple internalizing disorders. J Abnorm Psychol. 2017; 126(1): 8.Google Scholar
Alexopoulos, GS. The vascular depression hypothesis: 10 years later. Vol. 60, Biological Psychiatry . United States; 2006. pp. 13041305.Google Scholar
Fissler, M, Winnebeck, E, Schroeter, TA, et al. Brief training in mindfulness may normalize a blunted error-related negativity in chronically depressed patients. Cogn Affect Behav Neurosci. 2017 December; 17(6): 11641175.Google Scholar
Hegerl, U, Juckel, G. Intensity dependence of auditory evoked potentials as an indicator of central serotonergic neurotransmission: A new hypothesis* 1. Biological Psychiatry. 1993; 33: 173187.Google Scholar
Juckel, G, Molnar, M, Hegerl, U, Csepe, V, Karmos, G. Auditory-evoked potentials as indicator of brain serotonergic activity–first evidence in behaving cats. Biol Psychiatry. 1997 June; 41(12): 11811195.CrossRefGoogle ScholarPubMed
Juckel, G, Hegerl, U, Molnar, M, Csepe, V, Karmos, G. Auditory evoked potentials reflect serotonergic neuronal activity–a study in behaving cats administered drugs acting on 5-HT1A autoreceptors in the dorsal raphe nucleus. Neuropsychopharmacology. 1999 December; 21(6): 710716.Google Scholar
Jacobs, BL, Wilkinson, LO, Fornal, CA. The role of brain serotonin: A neurophysiologic perspective. Neuropsychopharmacology. 1990; 3(5–6): 473479.Google Scholar
Aghajanian, GK, Vandermaelen, CP. Specific Systems of the Reticular Core: Serotonin [Internet]. Comprehensive Physiology. 2011. (Major Reference Works). Available from: https://doi.org/10.1002/cphy.cp010404Google Scholar
Lewis, DA, Campbell, MJ, Foote, SL, Morrison, JH. The monoaminergic innervation of primate neocortex. Hum Neurobiol [Internet]. 1986; 5(3): 181188. Available from: http://europepmc.org/abstract/MED/3533864Google Scholar
Wutzler, A, Winter, C, Kitzrow, W, et al. Loudness dependence of auditory evoked potentials as indicator of central serotonergic neurotransmission: Simultaneous electrophysiological recordings and in vivo microdialysis in the rat primary auditory cortex. Neuropsychopharmacology [Internet]. 2008 May 7; 33: 3176. Available from: http://dx.doi.org/10.1038/npp.2008.42Google Scholar
Ruohonen, EM, Astikainen, P. Brain responses to sound intensity changes dissociate depressed participants and healthy controls. Biol Psychol [Internet]. 2017; 127: 7481. Available from: www.sciencedirect.com/science/article/pii/S0301051117301060Google Scholar
Gopal, K V, Bishop, CE, Carney, L. Auditory measures in clinically depressed individuals. II. Auditory evoked potentials and behavioral speech tests. Int J Audiol. 2004 October; 43(9): 499505.Google Scholar
Linka, T, Sartory, G, Bender, S, Gastpar, M, Müller, BW. The intensity dependence of auditory ERP components in unmedicated patients with major depression and healthy controls. An analysis of group differences. J Affect Disord [Internet]. 2007; 103(1): 139145. Available from: www.sciencedirect.com/science/article/pii/S0165032707000201Google Scholar
Park, Y-M, Lee, S-H, Kim, S, Bae, S-M. The loudness dependence of the auditory evoked potential (LDAEP) in schizophrenia, bipolar disorder, major depressive disorder, anxiety disorder, and healthy controls. Prog Neuro-Psychopharmacology Biol Psychiatry. 2010; 34(2): 313316.Google Scholar
Chen, T-J, Yu, YW-Y, Chen, M-C, et al. Serotonin dysfunction and suicide attempts in major depressives: An auditory event-related potential study. Neuropsychobiology. 2005; 52(1): 2836.Google Scholar
Fitzgerald, PB, Mellow, TB, Hoy, KE, et al. A study of intensity dependence of the auditory evoked potential (IDAEP) in medicated melancholic and non-melancholic depression. J Affect Disord. 2009; 117(3): 212216.Google Scholar
Jaworska, N, Blier, P, Fusee, W, Knott, V. Scalp- and sLORETA-derived loudness dependence of auditory evoked potentials (LDAEPs) in unmedicated depressed males and females and healthy controls. Clin Neurophysiol [Internet]. 2012 September; 123(9): 17691778. Available from: http://dx.doi.org/10.1016/j.clinph.2012.02.076Google Scholar
Lee, B-H, Park, Y-M, Lee, S-H, Shim, M. Prediction of long-term treatment response to selective serotonin reuptake inhibitors (SSRIs) using scalp and source loudness dependence of auditory evoked potentials (LDAEP) Analysis in patients with major depressive disorder. Int J Mol Sci [Internet]. 2015; 16(3): 62516265. Available from: www.mdpi.com/1422–0067/16/3/6251Google Scholar
Mulert, C, Juckel, G, Augustin, H, Hegerl, U. Comparison between the analysis of the loudness dependency of the auditory N1/P2 component with LORETA and dipole source analysis in the prediction of treatment response to the selective serotonin reuptake inhibitor citalopram in major depression. Clin Neurophysiol [Internet]. 2002; 113(10): 15661572. Available from: www.sciencedirect.com/science/article/pii/S1388245702002523Google Scholar
Jaworska, N, Blondeau, C, Tessier, P, et al. Response prediction to antidepressants using scalp and source-localized loudness dependence of auditory evoked potential (LDAEP) slopes. Prog Neuro-Psychopharmacology Biol Psychiatry [Internet]. 2013; 44: 100107. Available from: www.sciencedirect.com/science/article/pii/S0278584613000146Google Scholar
Gallinat, J, Bottlender, R, Juckel, G, et al. The loudness dependency of the auditory evoked N1/P2-component as a predictor of the acute SSRI response in depression. Psychopharmacology (Berl) [Internet]. 2000 March; 148(4): 404411. Available from: https://doi.org/10.1007/s002130050070Google Scholar
Gur, E, Lerer, B, Dremencov, E, Newman, ME. Chronic repetitive transcranial magnetic stimulation induces subsensitivity of presynaptic serotonergic autoreceptor activity in rat brain. Neuroreport [Internet]. 2000; 11(13). Available from: https://journals.lww.com/neuroreport/Fulltext/2000/09110/Chronic_repetitive_transcranial_magnetic.19.aspxGoogle Scholar
Gur, E, Lerer, B, Van de Kar, LD, Newman, ME. Chronic rTMS induces subsensitivity of post-synaptic 5-HT1A receptors in rat hypothalamus. Int J Neuropsychopharmacol [Internet]. 2004 September 1; 7(3): 335340. Available from: http://dx.doi.org/10.1017/S1461145703003985Google Scholar
Sibon, I, Strafella, AP, Gravel, P, et al. Acute prefrontal cortex TMS in healthy volunteers: Effects on brain 11 C-αMtrp trapping. Neuroimage [Internet]. 2007; 34(4): 16581664. Available from: www.sciencedirect.com/science/article/pii/S1053811906008846Google Scholar
Lee, S, Jang, K-I, Chae, J-H. Association of the loudness dependence of auditory evoked potentials with clinical changes to repetitive transcranial magnetic stimulation in patients with depression. J Affect Disord. 2018 October; 238: 451457.Google Scholar
Fox, MD, Buckner, RL, White, MP, Greicius, MD, Pascual-Leone, A. Efficacy of transcranial magnetic stimulation targets for depression is related to intrinsic functional connectivity with the subgenual cingulate. Biol Psychiatry. 2012 October; 72(7): 595603.Google Scholar
Korb, AS, Cook, IA, Hunter, AM, Leuchter, AF. Brain electrical source differences between depressed subjects and healthy controls. Brain Topogr. 2008; 21(2): 138146.Google Scholar
Pollock, VE, Schneider, LS. Topographic quantitative EEG in elderly subjects with major depression. Psychophysiology [Internet]. 1990 July 1; 27(4): 438444. Available from: https://doi.org/10.1111/j.1469–8986.1990.tb02340.xGoogle Scholar
Lopes da Silva, F., Vos, J., Mooibroek, J, Rotterdam, A van. Relative contributions of intracortical and thalamo-cortical processes in the generation of alpha rhythms, revealed by partial coherence analysis. Electroencephalogr Clin Neurophysiol. 1980; 50(5–6): 449456.Google Scholar
Suffczynski, P. Computational model of thalamo-cortical networks: dynamical control of alpha rhythms in relation to focal attention. Int J Psychophysiol. 2001; 43(1): 2540.Google Scholar
Chaumon, M, Busch, NA. Prestimulus neural oscillations inhibit visual perception via modulation of response gain. J Cogn Neurosci [Internet]. 2014 April 17; 26(11): 25142529. Available from: https://doi.org/10.1162/jocn_a_00653Google Scholar
Laufs, H, Krakow, K, Sterzer, P, et al. Electroencephalographic signatures of attentional and cognitive default modes in spontaneous brain activity fluctuations at rest. 2003;(October) PNAS September 16, 2003 100 (19) 11053–11058.Google Scholar
Gonçalves, SI. Correlating the alpha rhythm to BOLD using simultaneous EEG/fMRI: inter-subject variability. NeuroImage (Orlando, Fla). 2006; 30(1): 203213.Google Scholar
Laufs, H, Holt, J, Elfont, R, et al. Where the BOLD signal goes when alpha EEG leaves. NeuroImage (Orlando, Fla). 2006; 31(4): 14081418.Google Scholar
de Munck, JC. The hemodynamic response of the alpha rhythm: An EEG/fMRI study. NeuroImage (Orlando, Fla). 2007; 35(3): 11421151.Google ScholarPubMed
Klimesch, W, Sauseng, P, Hanslmayr, S. EEG alpha oscillations : The inhibition – timing hypothesis. 2006; 3. Brain Research Reviews. Volume 53, Issue 1, January 2007, Pages 63–88.Google Scholar
Näpflin, M, Wildi, M, Sarnthein, J. Test-retest reliability of resting EEG spectra validates a statistical signature of persons. Vol. 118, Clinical Neurophysiology. Sarnthein, Johannes: Universitatsspital Zurich, Zurich, Switzerland, CH-8091, : Elsevier Science; 2007. pp. 25192524.Google Scholar
Begić, DD, Mahnik-Miloš, M, Grubišin, J. EEG characteristics in depression, “negative” and “positive” schizophrena. Psychiatr Danub. 2009; 21(4): 579584.Google Scholar
Carlsson, A. The dopamine theory revisited. Schizophrenia. 1995; 379400.Google Scholar
Olbrich, S, Van Dinteren, R, Arns, M. Personalized medicine: Review and perspectives of promising baseline EEG biomarkers in major depressive disorder and attention deficit hyperactivity disorder. Neuropsychobiology. 2016; 72(3–4): 229240.Google Scholar
Roemer, RA, Shagass, C, Dubin, W, Jaffe, R, Siegal, L. Quantitative EEG in elderly depressives. Brain Topogr. 1992; 4(4): 285290.Google Scholar
Begic, D, Popovic-Knapic, V, Grubisin, J, et al. Quantitative electroencephalography in schizophrenia and depression. Psychiatr Danub. 2011 December; 23(4): 355362.Google Scholar
Jaworska, N, Blier, P, Fusee, W, Knott, V. α Power, α asymmetry and anterior cingulate cortex activity in depressed males and females. J Psychiatr Res [Internet]. 2012; 46(11): 14831491. Available from: www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3463760&tool=pmcentrez&rendertype=abstractCrossRefGoogle ScholarPubMed
Grin-Yatsenko, V a, Baas, I, Ponomarev, V a, Kropotov, JD. EEG power spectra at early stages of depressive disorders. J Clin Neurophysiol. 2009; 26(6): 401406.Google Scholar
Bruder, GE, Sedoruk, JP, Stewart, JW, et al. Electroencephalographic alpha measures predict therapeutic response to a selective serotonin reuptake inhibitor antidepressant: Pre- and post-treatment findings. Biol Psychiatry. 2008; 63(12): 11711177.CrossRefGoogle ScholarPubMed
Davidson, RJ. Cerebral asymmetry, emotion, and affective style. In: Brain asymmetry. Cambridge, MA, US: The MIT Press; 1995. pp. 361387.Google Scholar
Davidson, RJ, Henriques, J. Regional brain function in sadness and depression. In: The Neuropsychology of Emotion. New York, NY, US: Oxford University Press; 2000. pp. 269297. (Series in affective science.).Google Scholar
Davidson, RJ, Lewis, DA, Alloy, LB, et al. Neural and behavioral substrates of mood and mood regulation. Biol Psychiatry [Internet]. 2002; 52(6): 478502. Available from: www.sciencedirect.com/science/article/pii/S0006322302014580Google Scholar
Tomarkenand, AJ, Keener, AD. Frontal brain asymmetry and depression: A self-regulatory perspective. Cogn Emot [Internet]. 1998 May 1; 12(3): 387420. Available from: https://doi.org/10.1080/026999398379655Google Scholar
Micoulaud-Franchi, J-A, Richieri, R, Cermolacce, M, et al. Parieto-temporal alpha EEG band power at baseline as a predictor of antidepressant treatment response with repetitive transcranial magnetic stimulation: A preliminary study. J Affect Disord [Internet]. 2012; 137(1): 156160. Available from: www.sciencedirect.com/science/article/pii/S0165032711007932Google Scholar
Chang, JS, Yoo, CS, Yi, SH, et al. An integrative assessment of the psychophysiologic alterations in young women with recurrent major depressive disorder. Psychosom Med. 2012 June; 74(5): 495500.Google Scholar
Henriques, JB, Davidson, RJ. Left frontal hypoactivation in depression. J Abnorm Psychol. 1991 November; 100(4): 535545.Google Scholar
Schaffer, CE, Davidson, RJ, Saron, C. Frontal and parietal electroencephalogram asymmetry in depressed and nondepressed subjects. Biol Psychiatry. 1983 July; 18(7): 753762.Google Scholar
Kemp, AH, Griffiths, K, Felmingham, KL, et al. Disorder specificity despite comorbidity: Resting EEG alpha asymmetry in major depressive disorder and post-traumatic stress disorder. Biol Psychol [Internet]. 2010; 85(2): 350354. Available from: http://dx.doi.org/10.1016/j.biopsycho.2010.08.001Google Scholar
Beeney, JE, Levy, KN, Gatzke-Kopp, LM, Hallquist, MN. EEG asymmetry in borderline personality disorder and depression following rejection. Personal Disord Theory, Res Treat. 2014; 5(2): 178185.Google Scholar
Gollan, JK, Hoxha, D, Chihade, D, et al. Frontal alpha EEG asymmetry before and after behavioral activation treatment for depression. Biol Psychol [Internet]. 2014; 99: 198208. Available from: www.sciencedirect.com/science/article/pii/S0301051114000623CrossRefGoogle ScholarPubMed
Olbrich, S, Arns, M. EEG biomarkers in major depressive disorder: Discriminative power and prediction of treatment response. Int Rev Psychiatry. 2013; 25(5): 604618.CrossRefGoogle ScholarPubMed
van der Vinne, N, Vollebregt, MA, van Putten, MJAM, Arns, M. Frontal alpha asymmetry as a diagnostic marker in depression: Fact or fiction? A meta-analysis. NeuroImage Clin. 2017; 16(July): 7987.Google Scholar
Davidson, RJ. Affect, cognition, and hemispheric specialization. In: Emotions, Cognition, and Behavior. New York, NY, US: Cambridge University Press; 1985. pp. 320365.Google Scholar
Kelley, NJ, Hortensius, R, Schutter, DJLG, Harmon-Jones, E. The relationship of approach/avoidance motivation and asymmetric frontal cortical activity: A review of studies manipulating frontal asymmetry. Int J Psychophysiol [Internet]. 2017; 119: 1930. Available from: www.sciencedirect.com/science/article/pii/S0167876017301770Google Scholar
Debener, S, Beauducel, A, Nessler, D, et al. Is resting anterior EEG alpha asymmetry a trait marker for depression? Findings for healthy adults and clinically depressed patients. Neuropsychobiology. 2000; 41(1): 3137.CrossRefGoogle ScholarPubMed
Pizzagalli, DA, Nitschke, JB, Oakes, TR, et al. Brain electrical tomography in depression: the importance of symptom severity, anxiety, and melancholic features. Biol Psychiatry [Internet]. 2002; 52(2): 7385. Available from: www.sciencedirect.com/science/article/pii/S0006322302013136Google Scholar
Allen, JJB, Coan, JA, Nazarian, M. Issues and assumptions on the road from raw signals to metrics of frontal EEG asymmetry in emotion. Biol Psychol [Internet]. 2004; 67(1): 183218. Available from: www.sciencedirect.com/science/article/pii/S0301051104000377Google Scholar
Stewart, JL, Coan, JA, Towers, DN, Allen, JJB. Frontal EEG asymmetry during emotional challenge differentiates individuals with and without lifetime major depressive disorder. J Affect Disord [Internet]. 2011; 129(1–3): 167174. Available from: http://dx.doi.org/10.1016/j.jad.2010.08.029Google Scholar
Thibodeau, R, Jorgensen, RS, Kim, S. Depression, anxiety, and resting frontal EEG asymmetry: A meta-analytic review. J Abnorm Psychol. 2006; 115(4): 715729.Google Scholar
Arns, M, Bruder, G, Hegerl, U, et al. EEG alpha asymmetry as a gender-specific predictor of outcome to acute treatment with different antidepressant medications in the randomized iSPOT-D study. Clin Neurophysiol [Internet]. 2016; 127(1): 509519. Available from: http://dx.doi.org/10.1016/j.clinph.2015.05.032Google Scholar
Carvalho, A, Moraes, H, Silveira, H, et al. EEG frontal asymmetry in the depressed and remitted elderly: Is it related to the trait or to the state of depression? J Affect Disord [Internet]. 2011; 129(1): 143148. Available from: www.sciencedirect.com/science/article/pii/S0165032710005598Google Scholar
Gold, C, Fachner, J, Erkkilä, J. Validity and reliability of electroencephalographic frontal alpha asymmetry and frontal midline theta as biomarkers for depression. Scand J Psychol [Internet]. 2013 April 1; 54(2): 118126. Available from: https://doi.org/10.1111/sjop.12022Google Scholar
Reid, SA, Duke, LM, Allen, JJB. Resting frontal electroencephalographic asymmetry in depression: Inconsistencies suggest the need to identify mediating factors. Psychophysiology [Internet]. 1998 July 1; 35(4): 389404. Available from: https://doi.org/10.1111/1469–8986.3540389Google Scholar
Segrave, RA, Cooper, NR, Thomson, RH, et al. Individualized Alpha Activity and Frontal Asymmetry in Major Depression. Clin EEG Neurosci [Internet]. 2011 January 1; 42(1): 4552. Available from: https://doi.org/10.1177/155005941104200110Google Scholar
Kentgen, M. L, Tenke, C, Pine, D, et al. Electroencephalographic asymmetries in adolescents with major depression: Influence of comorbidity with anxiety disorders. Journal of Abnormal Psychology. 2000; 109: 797802.Google Scholar
Knott, V, Mahoney, C, Kennedy, S, Evans, K. EEG power, frequency, asymmetry and coherence in male depression. Psychiatry Res Neuroimaging [Internet]. 2001; 106(2): 123140. Available from: www.sciencedirect.com/science/article/pii/S0925492700000809Google Scholar
Deldin, PJ, Chiu, P. Cognitive restructuring and EEG in major depression. Biol Psychol [Internet]. 2005; 70(3): 141151. Available from: www.sciencedirect.com/science/article/pii/S030105110500027XGoogle Scholar
Mathersul, D, Williams, LM, Hopkinson, PJ, Kemp, AH. Investigating models of affect: Relationships among EEG alpha asymmetry, depression, and anxiety. Vol. 8, Emotion. Kemp, Andrew H.: The Brain Dynamics Centre, Westmead Hospital, Acacia House, Hawkesbury Road, Westmead, NSW, Australia, 2145, : American Psychological Association; 2008. 560572.Google Scholar
Quraan, MA, Protzner, AB, Daskalakis, ZJ, et al. EEG power asymmetry and functional connectivity as a marker of treatment effectiveness in DBS surgery for depression. Neuropsychopharmacology [Internet]. 2013 November 28; 39: 1270. Available from: https://doi.org/10.1038/npp.2013.330Google Scholar
Price, GW, Lee, JW, Garvey, C, Gibson, N. Appraisal of sessional EEG features as a correlate of clinical changes in an rTMS treatment of depression. Clin EEG Neurosci [Internet]. 2008 July 1; 39(3): 131138. Available from: https://doi.org/10.1177/155005940803900307Google Scholar
Allen, JJB, Kline, JP. Frontal EEG asymmetry, emotion, and psychopathology: The first, and the next 25 years. Biol Psychol [Internet]. 2004; 67(1): 15. Available from: www.sciencedirect.com/science/article/pii/S0301051104000304CrossRefGoogle ScholarPubMed
Kaiser, AK, Doppelmayr, M, Iglseder, B. Electroencephalogram alpha asymmetry in geriatric depression. Z Gerontol Geriatr [Internet]. 2018; 51(2): 200205. Available from: https://doi.org/10.1007/s00391-016–1108-zGoogle Scholar
Bruder, GE, Stewart, JW, Tenke, CE, et al. Electroencephalographic and perceptual asymmetry differences between responders and nonresponders to an SSRI antidepressant. Biol Psychiatry. 2001; 49(5): 416425.CrossRefGoogle Scholar
Knott, V, Mahoney, C, Kennedy, S, Evans, K. Pre-treatment EEG and it’s relationship to depression severity and paroxetine treatment outcome. Pharmacopsychiatry. 2000; 33: 201205.Google Scholar
Ulrich, G, Haug, H-J, Stieglietz, R-D, Fahndrich, E. EEG characteristics of clinically defined on-drug-responders and non-responders – A comparison clomipramine vs. maprotiline. Pharmacopsychiatry [Internet]. 1988; 21(6): 367368. Available from: www.scopus.com/inward/record.uri?eid=2-s2.0–0024213743&partnerID=40&md5=73803912ace9ccfcd8af4ee36646e933CrossRefGoogle ScholarPubMed
Baskaran, A, Farzan, F, Milev, R, et al. The comparative effectiveness of electroencephalographic indices in predicting response to escitalopram therapy in depression: A pilot study. J Affect Disord [Internet]. 2017; 227(October 2017): 542549. Available from: https://doi.org/10.1016/j.jad.2017.10.028Google Scholar
Knott, VJ, I. Telner, J, D. Lapierre, Y, et al. Quantitative EEG in the prediction of antidepressant response to imipramine. J Affect Disord. 1996 August 1; 39(3): 175184.Google Scholar
Baskaran, A, Milev, R, McIntyre, RS. The neurobiology of the EEG biomarker as a predictor of treatment response in depression. Neuropharmacology. 2012; 63(4): 507513.Google Scholar
Heller, W, A. Etienne, M, A. Miller, G. Patterns of perceptual asymmetry in depression and anxiety: Implications for neuropsychological models of emotion and psychopathology. Journal of Abnormal Psychology. 1995; 104: 327333.Google Scholar
Arns, M, Spronk, D, Fitzgerald, PB. Potential differential effects of 9 Hz rTMS and 10 Hz rTMS in the treatment of depression. Brain Stimul. 2010; 3(2): 124126.Google Scholar
Bench, CJ, Friston, KJ, Brown, RG, et al. The anatomy of melancholia–focal abnormalities of cerebral blood flow in major depression. Psychol Med [Internet]. 1992; 22(3): 607615. Available from: www.ncbi.nlm.nih.gov/pubmed/1410086CrossRefGoogle ScholarPubMed
Drevets, WC, Videen, TO, Price, JL, et al. A functional anatomical study of unipolar depression. J Neurosci. 1992 September; 12(9): 36283641.Google Scholar
Mayberg, HS, Lewis, PJ, Regenold, W, Wagner, HNJ. Paralimbic hypoperfusion in unipolar depression. J Nucl Med. 1994 June; 35(6): 929934.Google Scholar
Seminowicz, DA, Mayberg, HS, McIntosh, AR, et al. Limbic-frontal circuitry in major depression: a path modeling metanalysis. Neuroimage. 2004 May; 22(1): 409418.Google Scholar
Klimesch, W. EEG alpha and theta oscillations reflect cognitive and memory performance: a review and analysis. Brain Res Rev [Internet]. 1999 April [cited 2014 August 15]; 29(2–3): 169195. Available from: www.sciencedirect.com/science/article/pii/S0165017398000563Google Scholar
Dharmadhikari, AS, Tandle, AL, Jaiswal, S V, et al. Frontal theta asymmetry as a biomarker of depression. East Asian Arch Psychiatry [Internet]. 2018; 28(1): 1722. Available from: www.easap.asia/index.php/find-issues/current-issue/item/795–1803-v28n1-p17Google Scholar
Kempermann, G, Kronenberg, G. Depressed new neurons – Adult hippocampal neurogenesis and a cellular plasticity hypothesis of major depression. Biol Psychiatry. 2003; 54(5): 499503.Google Scholar
Fingelkurts, AA, Fingelkurts, AA, Rytsälä, H, et al. Composition of brain oscillations in ongoing EEG during major depression disorder. Neurosci Res. 2006; 56(2): 133144.Google Scholar
Iosifescu, DV. Electroencephalography-derived biomarkers of antidepressant response. Harv Rev Psychiatry. 2011; 19(3): 144154.Google Scholar
Ishii, R, Shinosaki, K, Ukai, S, et al. Medial prefrontal cortex generates frontal midline theta rhythm. Neuroreport. 1999; 10(4): 675679.Google Scholar
Asada, H, Fukuda, Y, Tsunoda, S, Yamaguchi, M, Tonoike, M. Frontal midline theta rhythms reflect alternative activation of prefrontal cortex and anterior cingulate cortex in humans. Neurosci Lett. 1999; 274(1): 2932.Google Scholar
Pizzagalli, DA, Oakes, TR, Davidson, RJ. Coupling of theta activity and glucose metabolism in the human rostral anterior cingulate cortex: An EEG/PET study of normal and depressed subjects. Psychophysiology. 2003; 40(6): 939949.Google Scholar
Kwon, JS, Youn, T, Jung, HY. Right hemisphere abnormalities in major depression: Quantitative electroencephalographic findings before and after treatment. J Affect Disord. 1996; 40(3): 169173.Google Scholar
Nystrom, C, Matousek, M, Hallstrom, T. Relationships between EEG and clinical characteristics in major depressive disorder. Acta Psychiatr Scand. 1986; 73(4): 390394.CrossRefGoogle ScholarPubMed
Lieber, AL. Diagnosis and subtyping of depressive disorders by quantitative electroencephalography: II. Interhemispheric measures are abnormal in major depressives and frequency analysis may discriminate certain subtypes. The Hillside Journal of Clinical Psychiatry. 1988; 10: 8497.Google Scholar
Arns, M, Etkin, A, Hegerl, U, et al. Frontal and rostral anterior cingulate (rACC) theta EEG in depression: Implications for treatment outcome? Eur Neuropsychopharmacol [Internet]. 2015; 25(8): 11901200. Available from: http://dx.doi.org/10.1016/j.euroneuro.2015.03.007Google Scholar
Grin-Yatsenko, VA, Baas, I, Ponomarev, VA, Kropotov, JD. Independent component approach to the analysis of EEG recordings at early stages of depressive disorders. Clin Neurophysiol [Internet]. 2010; 121(3): 281289. Available from: http://dx.doi.org/10.1016/j.clinph.2009.11.015Google Scholar
Degabriele, R, Lagopoulos, J. A review of EEG and ERP studies in bipolar disorder. Acta Neuropsychiatr [Internet]. 2014/06/24. 2009; 21(2): 5866. Available from: www.cambridge.org/core/article/review-of-eeg-and-erp-studies-in-bipolar-disorder/C7825AAA07D3119CF55C54A4563ABCA2CrossRefGoogle Scholar
Mientus, S, Gallinat, J, Wuebben, Y, et al. Cortical hypoactivation during resting EEG in schizophrenics but not in depressives and schizotypal subjects as revealed by low resolution electromagnetic tomography (LORETA). Psychiatry Res – Neuroimaging. 2002; 116(1–2): 95111.Google Scholar
Lubar, JF, Congedo, M, Askew, JH. Low-resolution electromagnetic tomography (LORETA) of cerebral activity in chronic depressive disorder. Int J Psychophysiol. 2003 September; 49(3): 175185.Google Scholar
Heikman, P, Salmelin, R, Mäkelä, JP, et al. Relation between frontal 3–7 Hz MEG activity and the efficacy of ECT in major depression. J ECT. 2001; 17(2): 136140.Google Scholar
Iosifescu, D V., Greenwald, S, Devlin, P, et al. Frontal EEG predictors of treatment outcome in major depressive disorder. Eur Neuropsychopharmacol [Internet]. 2009; 19(11): 772777. Available from: http://dx.doi.org/10.1016/j.euroneuro.2009.06.001CrossRefGoogle ScholarPubMed
Pizzagalli, DA. Frontocingulate dysfunction in depression: Toward biomarkers of treatment response. Neuropsychopharmacology [Internet]. 2011; 36(1): 183206. Available from: http://dx.doi.org/10.1038/npp.2010.166Google Scholar
Spronk, D, Arns, M, Barnett, KJ, Cooper, NJ, Gordon, E. An investigation of EEG, genetic and cognitive markers of treatment response to antidepressant medication in patients with major depressive disorder: A pilot study. J Affect Disord [Internet]. 2011; 128(1–2): 4148. Available from: http://dx.doi.org/10.1016/j.jad.2010.06.021Google Scholar
Korb, AS, Hunter, AM, Cook, IA, Leuchter, AF. Rostral anterior cingulate cortex theta current density and response to antidepressants and placebo in major depression. Clin Neurophysiol [Internet]. 2009; 120(7): 13131319. Available from: http://dx.doi.org/10.1016/j.clinph.2009.05.008Google Scholar
Mulert, C, Juckel, G, Brunnmeier, M, et al. Prediction of treatment response in major depression: Integration of concepts. J Affect Disord. 2007; 98(3): 215225.Google Scholar
Pizzagalli, DA, Pascual-Marqui, RD, Nitschke, JB, et al. Anterior cingulate activity as a predictor of degree of treatment response in major depression: Evidence from brain electrical tomography analysis. Am J Psychiatry [Internet]. 2001; 158(3): 405415. Available from: http://ajp.psychiatryonline.org.myaccess.library.utoronto.ca/doi/abs/10.1176/appi.ajp.158.3.405%5Cnfiles/523/Pizzagalli et al. – 2001 – Anterior Cingulate Activity as a Predictor of Degr.pdfGoogle Scholar
Watson, BO, Ding, M, Buzsáki, G. Temporal coupling of field potentials and action potentials in the neocortex. Eur J Neurosci [Internet]. 2018 October 1; 48(7): 24822497. Available from: https://doi.org/10.1111/ejn.13807Google Scholar
Nir, Y, Fisch, L, Mukamel, R, et al. Coupling between neuronal firing rate, gamma LFP, and BOLD fMRI is related to interneuronal correlations. Curr Biol. 2007; 17(15): 12751285.Google Scholar
Fries, P, Reynolds, JH, Rorie, AE, Desimone, R. Modulation of oscillatory neuronal synchronization by selective visual attention. Science. 2001 February; 291(5508): 15601563.Google Scholar
Kim, H, Ährlund-Richter, S, Wang, X, Deisseroth, K, Carlén, M. Prefrontal parvalbumin neurons in control of attention. Cell. 2016; 164(1–2): 208218.Google Scholar
Fitzgerald, PJ, Watson, BO. Gamma oscillations as a biomarker for major depression: an emerging topic. Transl Psychiatry [Internet]. 2018; 8(1): 177. Available from: https://doi.org/10.1038/s41398-018–0239-yGoogle Scholar
Uhlhaas, PJ, Haenschel, C, Nikolić, D, Singer, W. The role of oscillations and synchrony in cortical networks and their putative relevance for the pathophysiology of schizophrenia. Schizophr Bull [Internet]. 2008 September [cited 2014 October 1]; 34(5): 927943. Available from: www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2632472&tool=pmcentrez&rendertype=abstractGoogle Scholar
Colgin, LL, Denninger, T, Fyhn, M, et al. Frequency of gamma oscillations routes flow of information in the hippocampus. Nature [Internet]. 2009 November 19; 462: 353. Available from: https://doi.org/10.1038/nature08573Google Scholar
Fernández-Ruiz, A, Oliva, A, Nagy, GA, et al. Entorhinal-CA3 dual-input control of spike timing in the hippocampus by theta-gamma coupling. Neuron [Internet]. 2017; 93(5): 1213–1226.e5. Available from: www.sciencedirect.com/science/article/pii/S0896627317301010Google Scholar
Spellman, T, Rigotti, M, Ahmari, SE, et al. Hippocampal–prefrontal input supports spatial encoding in working memory. Nature [Internet]. 2015 June 8; 522: 309. Available from: https://doi.org/10.1038/nature14445CrossRefGoogle ScholarPubMed
Strelets, VB, Garakh, Z V, Novototskiĭ-Vlasov, VI. Comparative study of the gamma-rhythm in the norm, pre-examination stress and patients with the first depressive episode. Zh Vyssh Nerv Deiat Im I P Pavlova [Internet]. 2006; 56(2): 219227. Available from: www.ncbi.nlm.nih.gov/pubmed/16756129Google Scholar
Lee, PS, Chen, YS, Hsieh, JC, Su, TP, Chen, LF. Distinct neuronal oscillatory responses between patients with bipolar and unipolar disorders: A magnetoencephalographic study. J Affect Disord [Internet]. 2010; 123(1–3): 270275. Available from: http://dx.doi.org/10.1016/j.jad.2009.08.020Google Scholar
Liu, TY, Chen, YS, Su, TP, Hsieh, JC, Chen, LF. Abnormal early gamma responses to emotional faces differentiate unipolar from bipolar disorder patients. Biomed Res Int. 2014; 2014.Google Scholar
Pizzagalli, DA, Peccoralo, LA, Davidson, RJ, Cohen, JD. Resting anterior cingulate activity and abnormal responses to errors in subjects with elevated depressive symptoms: A 128-channel EEG study. Hum Brain Mapp [Internet]. 2006 March 1; 27(3): 185201. Available from: https://doi.org/10.1002/hbm.20172Google Scholar
Isomura, S, Onitsuka, T, Tsuchimoto, R, et al. Differentiation between major depressive disorder and bipolar disorder by auditory steady-state responses. J Affect Disord. 2016; 190(2016): 800806.Google Scholar
Oda, Y, Onitsuka, T, Tsuchimoto, R, Hirano, S, Oribe, N, Ueno, T, et al. Gamma band neural synchronization deficits for auditory steady state responses in bipolar disorder patients. PLoS One [Internet]. 2012 July; 7(7): e39955. Available from: http://dx.doi.org/10.1371/journal.pone.0039955Google Scholar
Liu, T-Y, Hsieh, J-C, Chen, Y-S, et al. Different patterns of abnormal gamma oscillatory activity in unipolar and bipolar disorder patients during an implicit emotion task. Neuropsychologia [Internet]. 2012 June [cited 2014 October 3]; 50(7): 15141520. Available from: www.ncbi.nlm.nih.gov/pubmed/22406691Google Scholar
Berman, RM, Cappiello, A, Anand, A, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000 February; 47(4): 351354.Google Scholar
Hong, LE, Summerfelt, A, Buchanan, RW, et al. Gamma and delta neural oscillations and association with clinical symptoms under subanesthetic ketamine. Neuropsychopharmacology [Internet]. 2009 November 4; 35: 632. Available from: https://doi.org/10.1038/npp.2009.168CrossRefGoogle ScholarPubMed
Muthukumaraswamy, SD, Shaw, AD, Jackson, LE, et al. Evidence that subanesthetic doses of Ketamine cause sustained disruptions of NMDA and AMPA-mediated frontoparietal connectivity in humans. J Neurosci [Internet]. 2015 August 19; 35(33): 11694LP11706. Available from: www.jneurosci.org/content/35/33/11694.abstractGoogle Scholar
Shaw, AD, Saxena, N, Jackson, LE, et al. Ketamine amplifies induced gamma frequency oscillations in the human cerebral cortex. Eur Neuropsychopharmacol [Internet]. 2015; 25(8): 11361146. Available from: http://dx.doi.org/10.1016/j.euroneuro.2015.04.012Google Scholar
Nugent, AC, Ballard, ED, Gould, TD, et al. Ketamine has distinct electrophysiological and behavioral effects in depressed and healthy subjects. Mol Psychiatry [Internet]. 2018; Available from: https://doi.org/10.1038/s41380-018–0028-2Google Scholar
Noda, Y, Zomorrodi, R, Saeki, T, et al. Resting-state EEG gamma power and theta–gamma coupling enhancement following high-frequency left dorsolateral prefrontal rTMS in patients with depression. Clin Neurophysiol [Internet]. 2017; 128(3): 424432. Available from: http://dx.doi.org/10.1016/j.clinph.2016.12.023Google Scholar
Bailey, NW, Hoy, KE, Rogasch, NC, et al. Responders to rTMS for depression show increased fronto-midline theta and theta connectivity compared to non-responders. Brain Stimul [Internet]. 2018; 11(1): 190203. Available from: https://doi.org/10.1016/j.brs.2017.10.015Google Scholar
Pathak, Y, Salami, O, Baillet, S, Li, Z, Butson, CR. Longitudinal Changes in Depressive Circuitry in Response to Neuromodulation Therapy [Internet]. Vol. 10, Frontiers in Neural Circuits; 2016. p. 50. Available from: www.frontiersin.org/article/10.3389/fncir.2016.00050Google Scholar
Belmaker, RH, Agam, G. Major depressive disorder. N Engl J Med [Internet]. 2008 January 3; 358(1): 5568. Available from: https://doi.org/10.1056/NEJMra073096Google Scholar
Gandal, MJ, Haney, JR, Parikshak, NN, et al. Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap. Science (80-) [Internet]. 2018 February 9; 359(6376): 693LP697. Available from: http://science.sciencemag.org/content/359/6376/693.abstractGoogle Scholar
Drysdale, AT, Grosenick, L, Downar, J, et al. HHS Public Access. 2017; 23(1): 2838.Google Scholar

References

Hansen, PC, Kringelbach, ML, Salmelin, R, editors. MEG: An Introduction to Methods. Oxford: Oxford University Press; 2010.Google Scholar
Hari, R, Puce, A, editors. MEG-EEG Primer. Oxford: Oxford University Press; 2017.Google Scholar
Supek, S, Aine, CJ, editors. Magnetoencephalography: From Signals to Dynamic Cortical Networks. Berlin: Springer-Verlag; 2014.Google Scholar
Boto, E, Holmes, N, Leggett, J, et al. Moving magnetoencephalography towards real-world applications with a wearable system. Nature. 2018; 555(7698): 657661.Google Scholar
Buzsáki, G. Rhythms of the Brain. Oxford: Oxford University Press; 2006.Google Scholar
Salustri, C, Tecchio, F, Zappasodi, F, et al. Cortical excitability and rest activity properties in patients with depression. Journal of Psychiatry and Neuroscience. 2007; 32(4): 259266.Google Scholar
Kurita, S, Takei, Y, Maki, Y, et al. Magnetoencephalography study of the effect of attention modulation on somatosensory processing in patients with major depressive disorder. Psychiatry and Clinical Neurosciences. 2016; 70(2): 116125.Google Scholar
Cornwell, BR, Salvadore, G, Furey, M, et al. Synaptic potentiation is critical for rapid antidepressant response to ketamine in treatment-resistant major depression. Biological Psychiatry. 2012; 72(7): 555561.Google Scholar
Nugent, AC, Wills, KE, Gilbert, JR, Zarate, Jr CA. Synaptic Potentiation and Rapid Antidepressant Response to Ketamine in Treatment-Resistant Major Depression: A Replication Study. In Revision.Google Scholar
Gilbert, JR, Yarrington, JS, Wills, KE, Nugent, AC, Zarate, CA, Jr. Glutamatergic signaling drives ketamine-mediated response in depression: Evidence from dynamic causal modeling. Int J Neuropsychopharmacol. 2018; 21(8): 740747.Google Scholar
Wang, Y, Jia, Y, Feng, Y, et al. Overlapping auditory M100 and M200 abnormalities in schizophrenia and bipolar disorder: A MEG study. Schizophrenia Research. 2014; 160(1–3): 201207.Google Scholar
Kähkönen, S, Yamashita, H, Rytsälä, H, et al. Dysfunction in early auditory processing in major depressive disorder revealed by combined MEG and EEG. Journal of Psychiatry and Neuroscience. 2007; 32(5): 316–222.Google Scholar
Takei, Y, Kumano, S, Hattori, S, et al. Preattentive dysfunction in major depression: A magnetoencephalography study using auditory mismatch negativity. Psychophysiology. 2009; 46(1): 5261.Google Scholar
Takei, Y, Kumano, S, Maki, Y, et al. Preattentive dysfunction in bipolar disorder: A MEG study using auditory mismatch negativity. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2010; 34(6): 903912.Google Scholar
Wang, Y, Feng, Y, Jia, Y, et al. Absence of auditory M100 source asymmetry in schizophrenia and bipolar disorder: A MEG study. PLoS ONE. 2013; 8(12).Google ScholarPubMed
Hegerl, U, Juckel, G. Intensity dependence of auditory evoked potentials as an indicator of central serotonergic neurotransmission: A new hypothesis. Biol Psychiatry. 1993; 33(3): 173187.Google Scholar
Hirakawa, N, Hirano, Y, Nakamura, I, et al. Right hemisphere pitch-mismatch negativity reduction in patients with major depression: An MEG study. Journal of Affective Disorders. 2017; 215: 225229.Google Scholar
Shimano, S, Onitsuka, T, Oribe, N, et al. Preattentive dysfunction in patients with bipolar disorder as revealed by the pitch-mismatch negativity: A magnetoencephalography (MEG) study. Bipolar Disorders. 2014; 16(6): 592599.CrossRefGoogle ScholarPubMed
Isomura, S, Onitsuka, T, Tsuchimoto, R, et al. Differentiation between major depressive disorder and bipolar disorder by auditory steady-state responses. Journal of Affective Disorders. 2016; 190: 800806.Google Scholar
Oda, Y, Onitsuka, T, Tsuchimoto, R, et al. Gamma band neural synchronization deficits for auditory steady state responses in bipolar disorder patients. PLoS ONE. 2012; 7(7).Google Scholar
Hirano, Y, Oribe, N, Kanba, S, et al. Spontaneous gamma activity in schizophrenia. JAMA Psychiatry. 2015; 72(8): 813821.Google Scholar
Nugent, AC, Ballard, ED, Gould, TD, et al. Ketamine has distinct electrophysiological and behavioral effects in depressed and healthy subjects. Mol Psychiatry. 2018; 24(7): 10401052.Google Scholar
Noda, Y, Zomorrodi, R, Saeki, T, et al. Resting-state EEG gamma power and theta-gamma coupling enhancement following high-frequency left dorsolateral prefrontal rTMS in patients with depression. Clin Neurophysiol. 2017; 128(3): 424432.Google Scholar
Kam, JW, Bolbecker, AR, O’Donnell, BF, Hetrick, WP, Brenner, CA. Resting state EEG power and coherence abnormalities in bipolar disorder and schizophrenia. J Psychiatr Res. 2013; 47(12): 18931901.Google Scholar
Reite, M, Teale, P, Rojas, DC, et al. MEG auditory evoked fields suggest altered structural/functional asymmetry in primary but not secondary auditory cortex in bipolar disorder. Bipolar Disorders. 2009; 11(4): 371381.Google Scholar
Wang, Y, Feng, Y, Jia, Y, et al. Auditory M50 and M100 sensory gating deficits in bipolar disorder: A MEG study. Journal of Affective Disorders. 2014; 152–154(1): 131138.Google Scholar
Oribe, N, Onitsuka, T, Hirano, S, et al. Differentiation between bipolar disorder and schizophrenia revealed by neural oscillation to speech sounds: An MEG study. Bipolar Disorders. 2010; 12(8): 804812.Google Scholar
Domschke, K, Zwanzger, P, Rehbein, MA, et al. Magnetoencephalographic correlates of emotional processing in major depression before and after pharmacological treatment. International Journal of Neuropsychopharmacology. 2016; 19(2): 19.Google Scholar
Xu, Q, Ruohonen, EM, Ye, C, et al. Automatic processing of changes in facial emotions in dysphoria: A magnetoencephalography study. Frontiers in Human Neuroscience. 2018; 12.Google Scholar
Moratti, S, Rubio, G, Campo, P, Keil, A, Ortiz, T. Hypofunction of right temporoparietal cortex during emotional arousal in depression. Archives of General Psychiatry. 2008; 65(5): 532541.Google Scholar
Moratti, S, Strange, B, Rubio, G. Emotional arousal modulation of right temporoparietal cortex in depression depends on parental depression status in women: First evidence. Journal of Affective Disorders. 2015; 178: 7987.Google Scholar
Disner, SG, Beevers, CG, Haigh, EA, Beck, AT. Neural mechanisms of the cognitive model of depression. Nat Rev Neurosci. 2011; 12(8): 467477.Google Scholar
Salvadore, G, Cornwell, BR, Colon-Rosario, V, et al. Increased anterior cingulate cortical activity in response to fearful faces: A neurophysiological biomarker that predicts rapid antidepressant response to ketamine. Biological Psychiatry. 2009; 65(4): 289295.Google Scholar
Lu, Q, Bi, K, Liu, C, et al. Predicting depression based on dynamic regional connectivity: A windowed Granger causality analysis of MEG recordings. Brain Research. 2013; 1535: 5260.CrossRefGoogle ScholarPubMed
Lu, Q, Li, H, Luo, G, et al. Impaired prefrontal-amygdala effective connectivity is responsible for the dysfunction of emotion process in major depressive disorder: A dynamic causal modeling study on MEG. Neuroscience Letters. 2012; 523(2): 125130.Google Scholar
Lu, Q, Wang, Y, Luo, G, Li, H, Yao, Z. Dynamic connectivity laterality of the amygdala under negative stimulus in depression: A MEG study. Neuroscience Letters. 2013; 547: 4247.Google Scholar
Bi, K, Hua, L, Wei, M, et al. Dynamic functional-structural coupling within acute functional state change phases: Evidence from a depression recognition study. Journal of Affective Disorders. 2016; 191: 145155.Google Scholar
Lu, Q, Jiang, H, Luo, G, Han, Y, Yao, Z. Multichannel matching pursuit of MEG signals for discriminative oscillation pattern detection in depression. International Journal of Psychophysiology. 2013; 88(2): 206212.Google Scholar
Lee, PS, Chen, YS, Hsieh, JC, Su, TP, Chen, LF. Distinct neuronal oscillatory responses between patients with bipolar and unipolar disorders: A magnetoencephalographic study. Journal of Affective Disorders. 2010; 123(1–3): 270275.Google Scholar
Liu, TY, Hsieh, JC, Chen, YS, et al. Different patterns of abnormal gamma oscillatory activity in unipolar and bipolar disorder patients during an implicit emotion task. Neuropsychologia. 2012; 50(7): 15141520.Google Scholar
McKinnon, MC, Yucel, K, Nazarov, A, MacQueen, GM. A meta-analysis examining clinical predictors of hippocampal volume in patients with major depressive disorder. J Psychiatry Neurosci. 2009; 34(1): 4154.Google Scholar
Cornwell, BR, Salvadore, G, Colon-Rosario, V, et al. Abnormal hippocampal functioning and impaired spatial navigation in depressed individuals: Evidence from whole-head magnetoencephalography. American Journal of Psychiatry. 2010; 167(7): 836844.Google Scholar
Salvadore, G, Cornwell, BR, Sambataro, F, et al. Anterior cingulate desynchronization and functional connectivity with the amygdala during a working memory task predict rapid antidepressant response to ketamine. Neuropsychopharmacology. 2010; 35(7): 14151422.Google Scholar
Ionescu, DF, Nugent, AC, Luckenbaugh, DA, et al. Baseline working memory activation deficits in dimensional anxious depression as detected by magnetoencephalography. Acta Neuropsychiatrica. 2015; 27(3): 143152.Google Scholar
Fernández, A, Rodriguez-Palancas, A, López-Ibor, M, et al. Increased occipital delta dipole density in major depressive disorder determined by magnetoencephalography. Journal of Psychiatry and Neuroscience. 2005; 30(1): 1723.Google Scholar
Maihöfner, C, Ropohl, A, Reulbach, U, et al. Effects of repetitive transcranial magnetic stimulation in depression: A magnetoencephalographic study. NeuroReport. 2005; 16(16): 18391842.Google Scholar
Al-Timemy, AH, Fernandez, A, Escudero, J, editors. Spectral Analysis of Resting State Magnetoencephalogram Activity in Patients with Bipolar Disorder. 2014.CrossRefGoogle Scholar
Jiang, H, Popov, T, Jylänki, P, et al. Predictability of depression severity based on posterior alpha oscillations. Clinical Neurophysiology. 2016; 127(4): 21082114.CrossRefGoogle ScholarPubMed
Linkenkaer-Hansen, K, Monto, S, Rytsälä, H, et al. Breakdown of long-range temporal correlations in theta oscillations in patients with major depressive disorder. Journal of Neuroscience. 2005; 25(44): 1013110137.Google Scholar
Buzsaki, G, Wang, XJ. Mechanisms of gamma oscillations. Annu Rev Neurosci. 2012; 35: 203225.Google Scholar
Gandal, MJ, Sisti, J, Klook, K, et al. GABAB-mediated rescue of altered excitatory-inhibitory balance, gamma synchrony and behavioral deficits following constitutive NMDAR-hypofunction. Transl Psychiatry. 2012; 2: e142.Google Scholar
Duman, RS, Aghajanian, GK. Synaptic dysfunction in depression: Potential therapeutic targets. Science. 2012; 338(6103): 6872.Google Scholar
Bachmann, M, Paeske, L, Kalev, K, et al. Methods for classifying depression in single channel EEG using linear and nonlinear signal analysis. Comput Methods Programs Biomed. 2018; 155: 1117.Google Scholar
Strelets, VB, Garakh Zh, V, Novototskii-Vlasov, VY. Comparative study of the gamma rhythm in normal conditions, during examination stress, and in patients with first depressive episode. Neurosci Behav Physiol. 2007; 37(4): 387394.Google Scholar
Shaw, AD, Saxena, N, Jackson, LE, et al. Ketamine amplifies induced gamma frequency oscillations in the human cerebral cortex. European Neuropsychopharmacology. 2015; 25(8): 11361146.Google Scholar
Zanos, P, Moaddel, R, Morris, PJ, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016; 533(7604): 481486.CrossRefGoogle ScholarPubMed
Stam, CJ. Nonlinear dynamical analysis of EEG and MEG: review of an emerging field. Clin Neurophysiol. 2005; 116(10): 22662301.Google Scholar
Méndez, MA, Zuluaga, P, Hornero, R, et al. Complexity analysis of spontaneous brain activity: Effects of depression and antidepressant treatment. Journal of Psychopharmacology. 2012; 26(5): 636643.Google Scholar
Fernández, A, Al-Timemy, AH, Ferre, F, Rubio, G, Escudero, J. Complexity analysis of spontaneous brain activity in mood disorders: A magnetoencephalography study of bipolar disorder and major depression. Comprehensive Psychiatry. 2018; 84: 112117.Google Scholar
Brookes, MJ, Woolrich, MW, Barnes, GR. Measuring functional connectivity in MEG: A multivariate approach insensitive to linear source leakage. Neuroimage. 2012; 63(2): 910920.Google Scholar
Brookes, MJ, Tewarie, PK, Hunt, BAE, et al. A multi-layer network approach to MEG connectivity analysis. Neuroimage. 2016; 132: 425438.Google Scholar
Colclough, GL, Brookes, MJ, Smith, SM, Woolrich, MW. A symmetric multivariate leakage correction for MEG connectomes. Neuroimage. 2015; 117: 439448.Google Scholar
Chen, SS, Tu, PC, Su, TP, et al. Impaired frontal synchronization of spontaneous magnetoencephalographic activity in patients with bipolar disorder. Neuroscience Letters. 2008; 445(2): 174178.Google Scholar
Nugent, AC, Robinson, SE, Coppola, R, Furey, ML, Zarate, CA, Jr. Group differences in MEG-ICA derived resting state networks: Application to major depressive disorder. NeuroImage. 2015; 118: 112.Google Scholar
Muthukumaraswamy, SD, Shaw, AD, Jackson, LE, et al. Evidence that subanesthetic doses of ketamine cause sustained disruptions of NMDA and AMPA-mediated frontoparietal connectivity in humans. Journal of Neuroscience. 2015; 35(33): 1169411706.Google Scholar
Zhang, S, Tian, S, Chattun, MR, et al. A supplementary functional connectivity microstate attached to the default mode network in depression revealed by resting-state magnetoencephalography. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2018; 83: 7685.Google Scholar
Wise, T, Marwood, L, Perkins, AM, et al. Instability of default mode network connectivity in major depression: A two-sample confirmation study. Transl Psychiatry. 2017; 7(4): e1105.Google Scholar
Hamilton, JP, Farmer, M, Fogelman, P, Gotlib, IH. Depressive rumination, the default-mode network, and the dark matter of clinical neuroscience. Biol Psychiatry. 2015; 78(4): 224230.Google Scholar
Zhu, X, Zhu, Q, Shen, H, Liao, W, Yuan, F. Rumination and default mode network subsystems connectivity in first-episode, drug-naive young patients with major depressive disorder. Sci Rep. 2017; 7: 43105.Google Scholar
Perera, T, George, MS, Grammer, G, et al. The clinical TMS society consensus review and treatment recommendations for TMS therapy for major depressive disorder. Brain Stimul. 2016; 9(3): 336346.Google Scholar
Pathak, Y, Salami, O, Baillet, S, Li, Z, Butson, CR. Longitudinal changes in depressive circuitry in response to neuromodulation therapy. Frontiers in Neural Circuits. 2016; 10(July 2016).Google Scholar
Li, CT, Chen, LF, Tu, PC, et al. Impaired prefronto-thalamic functional connectivity as a key feature of treatment-resistant depression: A combined MEG, PET and rTMS study. PLoS ONE. 2013; 8(8).Google Scholar

References

Collins, FS, Varmus, H. A new initiative on precision medicine. New England Journal of Medicine. 2015; 372(9): 793795.Google Scholar
Insel, TR, Cuthbert, BN. Brain disorders? precisely. Science. 2015; 348(6234): 499500.Google Scholar
Mrazek, DA, Hornberger, JC, Anthony Altar, C, Degtiar, I. A Review of the clinical, economic, and societal burden of treatment-resistant depression: 1996–2013. Psychiatric Services. 2014; 65(8): 977987.Google Scholar
Wong, EHF, Yocca, F, Smith, MA, Lee, C-M. Challenges and opportunities for drug discovery in psychiatric disorders: The drug hunters’ perspective. Int J Neuropsychopharmacol. 2010 October; 13(9): 12691284.Google Scholar
Hofmann, SG, Asnaani, A, Vonk, IJJ, Sawyer, AT, Fang, A. The efficacy of cognitive behavioral therapy: A review of meta-analyses. Cognitive Therapy and Research. 2012; 36(5): 427440.CrossRefGoogle ScholarPubMed
Cuthbert, BN, Insel, TR. Toward the future of psychiatric diagnosis: The seven pillars of RDoC. BMC Med. 2013 May 14; 11: 126.Google Scholar
Bzdok, D, Meyer-Lindenberg, A. Machine learning for precision psychiatry: Opportunities and challenges. Biol Psychiatry Cogn Neurosci Neuroimaging. 2018 March; 3(3): 223–30.Google ScholarPubMed
Iniesta, R, Stahl, D, McGuffin, P. Machine learning, statistical learning and the future of biological research in psychiatry. Psychol Med. 2016 September; 46(12): 24552465.Google Scholar
Varoquaux, G, Thirion, B. How machine learning is shaping cognitive neuroimaging. Gigascience. 2014 November 17; 3: 28.CrossRefGoogle ScholarPubMed
Librenza-Garcia, D, Kotzian, BJ, Yang, J, et al. The impact of machine learning techniques in the study of bipolar disorder: A systematic review. Neuroscience & Biobehavioral Reviews. 2017; 80: 538554.Google Scholar
Mwangi, B, Matthews, K, Douglas Steele, J. Prediction of illness severity in patients with major depression using structural MR brain scans. Journal of Magnetic Resonance Imaging. 2012; 35(1): 6471.Google Scholar
Williams, LM. Defining biotypes for depression and anxiety based on large-scale circuit dysfunction: A theoretical review of the evidence and future directions for clinical translation. Depress Anxiety. 2017 January; 34(1): 924.Google Scholar
Maia, TV, Frank, MJ. From reinforcement learning models to psychiatric and neurological disorders. Nat Neurosci. 2011 February; 14(2): 154162.Google Scholar
Huys, QJ, Pizzagalli, DA, Bogdan, R, Dayan, P. Mapping anhedonia onto reinforcement learning: a behavioural meta-analysis. Biol Mood Anxiety Disord. 2013 June 19; 3(1): 12.Google Scholar
Mwangi, B, Tian, TS, Soares, JC. A review of feature reduction techniques in neuroimaging. Neuroinformatics. 2014 April; 12(2): 229244.Google Scholar
Hastie, T, Tibshirani, R, Wainwright, M. Statistical Learning with Sparsity: The Lasso and Generalizations. CRC Press; 2015. 367.Google Scholar
Cristianini, N, Shawe-Taylor, J. An Introduction to Support Vector Machines and Other Kernel-based Learning Methods. Cambridge University Press; 2000. 189.Google Scholar
Hastie, T, Tibshirani, R, Friedman, J. The Elements of Statistical Learning: Data Mining, Inference, and Prediction. Springer; 2009.CrossRefGoogle Scholar
Plis, SM, Hjelm, DR, Salakhutdinov, R, et al. Deep learning for neuroimaging: A validation study. Front Neurosci. 2014 August 20; 8: 229.Google Scholar
Deutsch, H-P. Principle component analysis. In: Derivatives and Internal Models. Palgrave Macmillan, London; 2002. pp. 539547. (Finance and Capital Markets Series).Google Scholar
Stone, JV. Independent Component Analysis: A Tutorial Introduction. MIT Press; 2004.CrossRefGoogle Scholar
Kruskal, JB. Multidimensional scaling by optimizing goodness of fit to a nonmetric hypothesis. Psychometrika. 1964; 29(1): 127.Google Scholar
Roweis, ST. Nonlinear dimensionality reduction by locally linear embedding. Science. 2000; 290(5500): 23232326.Google Scholar
Lee, DD, Seung, HS. Learning the parts of objects by non-negative matrix factorization. Nature. 1999 October 21; 401(6755): 788791.Google Scholar
van der Maaten, L, Hinton, G. Visualizing data using t-SNE. J Mach Learn Res. 2008 November 8; 9: 25792605.Google Scholar
Calhoun, VD, Liu, J, Adali, T. A review of group ICA for fMRI data and ICA for joint inference of imaging, genetic, and ERP data. NeuroImage. 2009; 45(1): S163S172.Google Scholar
Jolliffe, IT. Principal Component Analysis. Springer; 2002. (Springer Series in Statistics).Google Scholar
Hartigan, JA, Wong, MA. Algorithm, AS 136: A K-means clustering algorithm. Applied Statistics. 1979; 28(1): 100.Google Scholar
Cheng, Y. Mean shift, mode seeking, and clustering. IEEE Transactions on Pattern Analysis and Machine Intelligence. 1995; 17(8): 790799.Google Scholar
Johnson, SC. Hierarchical clustering schemes. Psychometrika. 1967; 32(3): 241254.Google Scholar
Marquand, AF, Wolfers, T, Dinga, R. Phenomapping: Methods and measures for deconstructing diagnosis in psychiatry. Personalized Psychiatry. 2019; 119134.Google Scholar
Zhou, X-H, Obuchowski, NA, McClish, DK. Statistical Methods in Diagnostic Medicine. Wiley; 2011. (Wiley Series in Probability and Statistics).Google Scholar
Rousseeuw, PJ. Silhouettes: A graphical aid to the interpretation and validation of cluster analysis. Journal of Computational and Applied Mathematics. 1987; 20: 5365.Google Scholar
Mwangi, B, Soares, JC, Hasan, KM. Visualization and unsupervised predictive clustering of high-dimensional multimodal neuroimaging data. J Neurosci Methods. 2014 October 30; 236: 1925.Google Scholar
Zhang, W, Xiao, Y, Sun, H, et al. Discrete patterns of cortical thickness in youth with bipolar disorder differentially predict treatment response to quetiapine but not lithium. Neuropsychopharmacology. 2018; 43(11): 22562263.Google Scholar
Marquand, AF, Wolfers, T, Mennes, M, Buitelaar, J, Beckmann, CF. Beyond lumping and splitting: A review of computational approaches for stratifying psychiatric disorders. Biol Psychiatry Cogn Neurosci Neuroimaging. 2016 September; 1(5): 433447.Google Scholar
Wu, M-J, Mwangi, B, Bauer, IE, et al. Identification and individualized prediction of clinical phenotypes in bipolar disorders using neurocognitive data, neuroimaging scans and machine learning. Neuroimage. 2017 January 15; 145(Pt B): 254264.Google Scholar
Dunn, JC. Well-separated clusters and optimal fuzzy partitions. Journal of Cybernetics. 1974; 4(1): 95104.CrossRefGoogle Scholar
Davies, DL, Bouldin, DW. A cluster separation measure. IEEE Transactions on Pattern Analysis and Machine Intelligence. 1979; PAMI-1(2): 224227.Google Scholar
Tibshirani, R, Walther, G, Hastie, T. Estimating the number of clusters in a data set via the gap statistic. Journal of the Royal Statistical Society: Series B (Statistical Methodology). 2001; 63(2): 411423.Google Scholar
Hubert, L, Schultz, J. Quadratic assignment as a general data analysis strategy. British Journal of Mathematical and Statistical Psychology. 1976; 29(2): 190241.Google Scholar
Günter, S, Bunke, H. Validation indices for graph clustering. Pattern Recognition Letters. 2003; 24(8): 11071113.Google Scholar
Bishop, CM. Pattern Recognition and Machine Learning. Springer Verlag; 2006.Google Scholar
Kim, Y-K, Na, K-S. Application of machine learning classification for structural brain MRI in mood disorders: Critical review from a clinical perspective. Prog Neuropsychopharmacol Biol Psychiatry. 2018 January 3; 80(Pt B): 7180.Google Scholar
Dusi, N, De Carlo, V, Delvecchio, G, Bellani, M, Soares, JC, Brambilla, P. MRI features of clinical outcome in bipolar disorder: A selected review. Journal of Affective Disorders. 2019; 243: 559563.Google Scholar
Yoshida, K, Shimizu, Y, Yoshimoto, J, et al. Prediction of clinical depression scores and detection of changes in whole-brain using resting-state functional MRI data with partial least squares regression. PLoS One. 2017 July 12; 12(7): e0179638.Google Scholar
Rosa, AR, Sánchez-Moreno, J, Martínez-Aran, A, et al. Validity and reliability of the Functioning Assessment Short Test (FAST) in bipolar disorder. Clinical Practice and Epidemiology in Mental Health. 2007; 3(1): 5.Google Scholar
Sartori, JM, Reckziegel, R, Passos, IC, et al. Volumetric brain magnetic resonance imaging predicts functioning in bipolar disorder: A machine learning approach. J Psychiatr Res. 2018 August; 103: 237243.Google Scholar
Han, LKM, Dinga, R, Hahn, T, et al. Brain Aging in Major Depressive Disorder: Results from the ENIGMA Major Depressive Disorder working group [Internet]. bioRxiv. 2019 [cited 2019 Sep 5]. p. 560623. Available from: http://dx.doi.org/10.1101/560623Google Scholar
Nunes, A, for the ENIGMA Bipolar Disorders Working Group, Schnack HG, Ching CRK, Agartz I, Akudjedu TN, et al. Using structural MRI to identify bipolar disorders – 13 site machine learning study in 3020 individuals from the ENIGMA Bipolar Disorders Working Group. Molecular Psychiatry [Internet]. 2018; Available from: http://dx.doi.org/10.1038/s41380-018–0228-9Google Scholar
Grisanzio, KA, Goldstein-Piekarski, AN, Wang, MY, et al. Transdiagnostic symptom clusters and associations with brain, behavior, and daily function in mood, anxiety, and trauma disorders. JAMA Psychiatry. 2018; 75(2): 201.Google Scholar
Dinga, R, Schmaal, L, Penninx, BWJH, et al. Evaluating the evidence for biotypes of depression: Methodological replication and extension of. Neuroimage Clin. 2019 March 27; 22: 101796.Google Scholar
Harmer, CJ, Duman, RS, Cowen, PJ. How do antidepressants work? New perspectives for refining future treatment approaches. Lancet Psychiatry. 2017 May; 4(5): 409418.Google Scholar
Rutledge, RB, Chekroud, AM, Huys, QJ. Machine learning and big data in psychiatry: toward clinical applications. Curr Opin Neurobiol. 2019 April; 55: 152159.Google Scholar
Webb, CA, Trivedi, MH, Cohen, ZD, et al. Personalized prediction of antidepressant v. placebo response: Evidence from the EMBARC study. Psychol Med. 2019 May; 49(7): 11181127.Google Scholar
Rush, AJ, John Rush, A, Fava, M, et al. Sequenced treatment alternatives to relieve depression (STAR*D): Rationale and design. Controlled Clinical Trials. 2004; 25(1): 119142.Google Scholar
Paul, R, Andlauer, TFM, Czamara, D, et al. Treatment response classes in major depressive disorder identified by model-based clustering and validated by clinical prediction models. Translational Psychiatry [Internet]. 2019; 9(1). Available from: http://dx.doi.org/10.1038/s41398-019–0524-4Google Scholar
Khodayari-Rostamabad, A, Reilly, JP, Hasey, GM, de Bruin, H, MacCrimmon, DJ. A machine learning approach using EEG data to predict response to SSRI treatment for major depressive disorder. Clinical Neurophysiology. 2013; 124(10): 19751985.Google Scholar
Iniesta, R, Malki, K, Maier, W, et al. Combining clinical variables to optimize prediction of antidepressant treatment outcomes. J Psychiatr Res. 2016 July; 78: 94102.Google Scholar
Kautzky, A, Dold, M, Bartova, L, et al. Refining prediction in treatment-resistant depression: Results of machine learning analyses in the TRD III sample. J Clin Psychiatry [Internet]. 2018; 79(1). Available from: http://dx.doi.org/10.4088/JCP.16m11385Google Scholar
Cao, B, Luo, Q, Fu, Y, et al. Predicting individual responses to the electroconvulsive therapy with hippocampal subfield volumes in major depression disorder. Sci Rep. 2018 April 3; 8(1): 5434.Google Scholar
Redlich, R, Opel, N, Grotegerd, D, et al. Prediction of individual response to electroconvulsive therapy via machine learning on structural magnetic resonance imaging data. JAMA Psychiatry. 2016; 73(6): 557.Google Scholar
Fleck, DE, Ernest, N, Adler, CM, et al. Prediction of lithium response in first-episode mania using the LITHium Intelligent Agent (LITHIA): Pilot data and proof-of-concept. Bipolar Disord. 2017 June; 19(4): 259272.Google Scholar
Tseng, H-H, Luo, Y, Cui, S, et al. Deep reinforcement learning for automated radiation adaptation in lung cancer. Med Phys. 2017 December; 44(12): 66906705.Google Scholar
Schofield, P, Crosland, A, Waheed, W, et al. Patients’ views of antidepressants: From first experiences to becoming expert. Br J Gen Pract. 2011 April; 61(585): 142148.Google Scholar
Kessler, RC, Warner, CH, Ivany, C, et al. Predicting suicides after psychiatric hospitalization in US army soldiers: the army study to assess risk and rEsilience in servicemembers (Army STARRS). JAMA Psychiatry. 2015 January; 72(1): 4957.Google Scholar
Tran, T, Luo, W, Phung, D, et al. Risk stratification using data from electronic medical records better predicts suicide risks than clinician assessments. BMC Psychiatry. 2014 Mar 14; 14: 76.Google Scholar
Passos, IC, Mwangi, B, Cao, B, et al. Identifying a clinical signature of suicidality among patients with mood disorders: A pilot study using a machine learning approach. Journal of Affective Disorders. 2016; 193: 109116.CrossRefGoogle ScholarPubMed
Castro, VM, Roberson, AM, McCoy, TH, Wiste, A, Cagan, A, Smoller, JW, et al. Stratifying risk for renal insufficiency among lithium-treated patients: An electronic health record study. Neuropsychopharmacology. 2016 March; 41(4): 11381143.Google Scholar
Mwangi, B, Wu, M-J, Cao, B, et al. Individualized prediction and clinical staging of bipolar disorders using neuroanatomical biomarkers. Biol Psychiatry Cogn Neurosci Neuroimaging. 2016 March 1; 1(2): 186194.Google Scholar
Cao, B, Passos, IC, Mwangi, B, et al. Hippocampal volume and verbal memory performance in late-stage bipolar disorder. J Psychiatr Res. 2016 February; 73: 102107.Google Scholar
Lavagnino, L, Cao, B, Mwangi, B, -J. et al. Changes in the corpus callosum in women with late-stage bipolar disorder. Acta Psychiatrica Scandinavica. 2015; 131(6): 458464.Google Scholar
Passos, IC, Mwangi, B, Vieta, E, Berk, M, Kapczinski, F. Areas of controversy in neuroprogression in bipolar disorder. Acta Psychiatr Scand. 2016 August; 134(2): 91103.Google Scholar
Kapczinski, NS, Mwangi, B, Cassidy, RM, et al. Neuroprogression and illness trajectories in bipolar disorder. Expert Review of Neurotherapeutics. 2017; 17(3): 277285.Google Scholar
Silva, RF, Plis, SM. How to Integrate Data from Multiple Biological Layers in Mental Health? [Internet]. Personalized Psychiatry. 2019. p. 135–59. Available from: http://dx.doi.org/10.1007/978–3-030–03553-2_8Google Scholar
Honeine, P, Richard, C. Solving the pre-image problem in kernel machines: A direct method. 2009 IEEE International Workshop on Machine Learning for Signal Processing [Internet]. 2009; Available from: http://dx.doi.org/10.1109/mlsp.2009.5306204Google Scholar
Jobin, A, Ienca, M, Vayena, E. The global landscape of AI ethics guidelines. Nature Machine Intelligence [Internet]. 2019; Available from: http://dx.doi.org/10.1038/s42256-019–0088-2Google Scholar
Varoquaux, G, Raamana, PR, Engemann, DA, et al. Assessing and tuning brain decoders: Cross-validation, caveats, and guidelines. Neuroimage. 2017 January 15; 145(Pt B): 166179.Google Scholar
Thompson, PM, Stein, JL, Medland, SE, et al. The ENIGMA Consortium: large-scale collaborative analyses of neuroimaging and genetic data. Brain Imaging Behav. 2014; 8(2): 153.Google Scholar
Zhu, D, Riedel, BC, Jahanshad, N, et al. Classification of major depressive disorder via multi-site weighted LASSO model. Medical Image Computing and Computer Assisted Intervention − MICCAI 2017. 2017; 159167.Google Scholar
Developing Software Precertification Program: A Working Model [Internet]. FDA U.S. Food & Drug Administration; 2018 June. Available from: www.fda.gov/media/113802/downloadGoogle Scholar
Horvitz, E, Mulligan, D. Data, privacy, and the greater good. Science. 2015; 349(6245): 253255.Google Scholar
Ohno-Machado, L, Bafna, V, Boxwala, AA, et al. iDASH: Integrating data for analysis, anonymization, and sharing. Journal of the American Medical Informatics Association. 2012; 19(2): 196201.Google Scholar
Peterson, K, Deeduvanu, R, Kanjamala, P, Boles, K. A blockchain-based approach to health information exchange networks. NIST Workshop Blockchain Healthcare. 2016 September; 1: 110.Google Scholar
Passos, IC, Ballester, P, Barros, RC, et al. Machine learning and big data analytics in bipolar disorder: A Position paper from the International Society for Bipolar Disorders (ISBD) Big Data Task Force. Bipolar Disorders [Internet]. 2019; Available from: http://dx.doi.org/10.1111/bdi.12828Google Scholar
Vayena, E, Blasimme, A, Glenn Cohen, I. Machine learning in medicine: Addressing ethical challenges. PLoS Med. 2018 November 6; 15(11): e1002689.Google Scholar
Gianfrancesco, MA, Tamang, S, Yazdany, J, Schmajuk, G. Potential biases in machine learning algorithms using electronic health record data. JAMA Internal Medicine. 2018; 178(11): 1544.Google Scholar
O’Neil, C. Weapons of Math Destruction: How Big Data Increases Inequality and Threatens Democracy. Crown Books; 2016.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×