Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-r6qrq Total loading time: 0 Render date: 2024-04-26T07:49:59.768Z Has data issue: false hasContentIssue false

Section 5 - Therapeutic Applications of Neuroimaging in Mood Disorders

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. 219 - 272
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

Ferrari, AJ, Stockings, E, Khoo, JP, Erskine, HE, Degenhardt, L, Vos, T, Whiteford, HA. The prevalence and burden of bipolar disorder: findings from the Global Burden of Disease Study 2013. Bipolar Disord. 2016 August; 18(5): 440450. DOI 10.1111/bdi.12423. PubMed PMID: 27566286.Google Scholar
Machado-Vieira, R, Manji, HK, Zarate, CA Jr: The role of lithium in the treatment of bipolar disorder: convergent evidence for neurotrophic effects as a unifying hypothesis. Bipolar Disord. 2009 June; 11 Suppl 2 : 92109.PMCID: 2800957.CrossRefGoogle ScholarPubMed
Suwalska, A, Sobieska, M, Rybakowski, JK. Serum brain-derived neurotrophic factor in euthymic bipolar patients on prophylactic lithium therapy. Neuropsychobiology. 2010; 62(4): 229234. Epub 2010 August 14. PMID: 20714172.CrossRefGoogle ScholarPubMed
Baldessarini, RJ, Tondo, L, Vázquez, GH. Pharmacological treatment of adult bipolar disorder. Mol Psychiatry. 2018 April 20. DOI 10.1038/s41380-018-0044-2. [Epub ahead of print] Review. PubMed PMID: 29679069.Google Scholar
Schaffer, A, Isometsä, ET, Tondo, L, et al. Epidemiology, neurobiology and pharmacological interventions related to suicide deaths and suicide attempts in bipolar disorder: Part I of a report of theInternational Society for Bipolar Disorders Task Force on Suicide in Bipolar Disorder. Aust N Z J Psychiatry. 2015 September; 49(9): 785802. DOI 10.1177/0004867415594427. Epub 2015 July 16. Review. PubMed PMID: 26185269; PubMed Central PMCID: PMC5116383.Google Scholar
Hafeman, DM, Chang, KD, Garrett, AS, Sanders, EM, Phillips, ML. Effects of medication on neuroimaging findings in bipolar disorder: an updated review. Bipolar Disord. 2012 June;14(4): 375410. DOI 10.1111/j.1399-5618.2012.01023.x. Review. PubMed PMID: 22631621.Google Scholar
Cousins, DA, Aribisala, B, Ferrier, IN, Blamire, AM. Lithium, gray matter, and magnetic resonance imaging signal. Biol Psychiatry. 2013; 73: 652657.Google Scholar
Bearden, CE, Thompson, PM, Dutton, RA, et al. Three-dimensional mapping of hippocampal anatomy in unmedicated and lithium-treated patients with bipolar disorder. Neuropsychopharmacology. 2008 May; 33(6): 12291238. Epub 2007 August 8. PubMed PMID: 17687266.Google Scholar
Foland, LC, Altshuler, LL, Sugar, CA, et al. Increased volume of the amygdala and hippocampus in bipolar patients treated with lithium. Neuroreport. 2008 January 22; 19(2): 221224. DOI 10.1097/WNR.0b013e3282f48108. PubMed PMID: 18185112; PubMed Central PMCID: PMC3299336.Google Scholar
Hajek, T, Bauer, M, Simhandl, C, et al. Neuroprotective effect of lithium on hippocampal volumes in bipolar disorder independent of long-term treatment response. Psychol Med. 2014 February; 44(3): 507517. DOI 10.1017/S0033291713001165. Epub 2013 May 31. PubMed PMID: 23721695.Google Scholar
Baykara, B, Inal-Emiroglu, N, Karabay, N, et.al Increased hippocampal volumes in lithium treated adolescents with bipolar disorders: A structural MRI study. J Affect Disord. 2012 May; 138(3): 433439. DOI 10.1016/j.jad.2011.12.047. Epub 2012 February 9.CrossRefGoogle ScholarPubMed
Hajek, T, Cullis, J, Novak, T, et al. Hippocampal volumes in bipolar disorders: Opposing effects of illness burden and lithium treatment. Bipolar Disord. 2012 May; 14(3): 261270. DOI 10.1111/j.1399-5618.2012.01013.x.Google Scholar
Hajek, T, Kopecek, M, Höschl, C, Alda, M. Smaller hippocampal volumes in patients with bipolar disorder are masked by exposure to lithium: a meta-analysis. J Psychiatry Neurosci. 2012 September; 37(5): 333343. DOI:10.1503/jpn.110143.Google Scholar
Yucel, K, Taylor, VH, McKinnon, MC, et al. Bilateral hippocampal volume increase in patients with bipolar disorder and short-term lithium treatment. Neuropsychopharmacology. 2008 January; 33(2): 361367. Epub 2007 April 4. PubMed PMID: 17406649.Google Scholar
Giakoumatos, CI, Nanda, P, Mathew, IT, et al. Effects of lithium on cortical thickness and hippocampal subfield volumes in psychotic bipolar disorder. J Psychiatr Res. 2015 February; 61: 180187. DOI 10.1016/j.jpsychires.2014.12.008. Epub 2014 December 23. PubMed PMID: 25563516; PubMed Central PMCID: PMC4859940.Google Scholar
Hartberg, CB, Jørgensen, KN, Haukvik, UK, et al. Lithium treatment and hippocampal subfields and amygdala volumes in bipolar disorder. Bipolar Disord. 2015 August; 17(5): 496506. DOI:10.1111/bdi.12295. Epub 2015 March 24. PubMed PMID: 25809287.Google Scholar
Bearden, CE, Thompson, PM, Dalwani, M, et al. Greater cortical gray matter density in lithium-treated patients with bipolar disorder. Biol Psychiatry. 2007 July 1; 62(1): 716. Epub 2007 January 19. PubMed PMID: 17240360; PubMed Central PMCID: PMC3586797.Google Scholar
Germaná, C, Kempton, MJ, Sarnicola, A, et al. The effects of lithium and anticonvulsants on brain structure in bipolar disorder. Acta Psychiatr Scand. 2010 December; 122(6): 481487. DOI 10.1111/j.1600-0447.2010.01582.x. PubMed PMID: 20560901.Google Scholar
López-Jaramillo, C, Vargas, C, Díaz-Zuluaga, AM, et al. Increased hippocampal, thalamus and amygdala volume in long-term lithium-treated bipolar I disorder patients compared with unmedicated patients and healthy subjects. Bipolar Disord. 2017 February; 19(1): 4149. DOI 10.1111/bdi.12467. Epub 2017 Feb 27. PubMed PMID: 28239952.CrossRefGoogle ScholarPubMed
Sassi, RB, Nicoletti, M, Brambilla, P, et al. Increased gray matter volume in lithium-treated bipolar disorder patients. Neurosci Lett. 2002 August 30; 329(2):243245. PubMed PMID: 12165422.Google Scholar
Berk, M, Dandash, O, Daglas, R, et al. Neuroprotection after a first episode of mania: a randomized controlled maintenance trial comparing the effects of lithium and quetiapine on grey and white matter volume. Transl Psychiatry. 2017 January 24; 7(1): e1011. DOI 10.1038/tp.2016.281. Erratum in: Transl Psychiatry. 2017 February 21;7(2):e1041. PubMed PMID: 28117843; PubMed Central PMCID: PMC5545739.CrossRefGoogle Scholar
Yucel, K, McKinnon, MC, Taylor, VH, et al. Bilateral hippocampal volume increases after long-term lithium treatment in patients with bipolar disorder: A longitudinal MRI study. Psychopharmacology (Berl). 2007 December; 195 (3): 357367. Epub 2007 August 20. PubMed PMID: 17705060.Google Scholar
Selek, S, Nicoletti, M, Zunta-Soares, GB, et al. A longitudinal study of fronto-limbic brain structures in patients with bipolar I disorder during lithium treatment. J Affect Disord. 2013 September 5; 150(2): 629633. DOI 10.1016/j.jad.2013.04.020. Epub 2013 June 10. PubMed PMID: 23764385.Google Scholar
Lyoo, IK, Dager, SR, Kim, JE, et al. Lithium-induced gray matter volume increase as a neural correlate of treatment response in bipolar disorder: A longitudinal brain imaging study. Neuropsychopharmacology. 2010 July; 35(8): 17431750. DOI 10.1038/npp.2010.41. Epub 2010 March 31. PubMed PMID: 20357761; PubMed Central PMCID: PMC3055479.Google Scholar
Moore, GJ, Bebchuk, JM, Wilds, IB, Chen, G, Manji, HK. Lithium-induced increase in human brain grey matter. Lancet. 2000 Oct 7;356(9237):1241–2. Erratum in: Lancet 2000 December 16;356(9247):2104.Menji HK [corrected to Manji HK]. PubMed PMID: 11072948.Google Scholar
Moore, GJ, Cortese, BM, Glitz, DA, et al. A longitudinal study of the effects of lithium treatment on prefrontal and subgenual prefrontal gray matter volume in treatment-responsive bipolar disorder patients. J Clin Psychiatry. 2009 April 21; 70(5): 699705. DOI:10.4088/JCP.07m03745. PubMed PMID: 19389332.Google Scholar
Monkul, ES, Matsuo, K, Nicoletti, MA, et al. Prefrontal gray matter increases in healthy individuals after lithium treatment: A voxel-based morphometry study. Neurosci Lett. 2007 December 11; 429(1): 711. Epub 2007 October 10. PubMed PMID: 17996370; PubMed Central PMCID: PMC2693231.Google Scholar
Phatak, P, Shaldivin, A, King, LS, Shapiro, P, Regenold, WT. Lithium and inositol: effects on brain water homeostasis in the rat, Psychopharmacology (Berl). 2006; 186: 4147.Google Scholar
Necus, JM, Sinha, N, Smith, FE, et al. White matter microstructural properties in bipolar disorder and its relationship to the spatial distribution of lithium in the brain. Submitted doi:http://dx.doi.org/10.1101/346528 bioRxiv preprint first posted online Jun. 13, 2018.Google Scholar
Riadh, N, Allagui, MS, Bourogaa, E, et al. Neuroprotective and neurotrophic effects of long term lithium treatment in mouse brain. Biometals. 2011; 24: 747757. DOI 10.1007/s10534-011–9433-6.Google Scholar
Vernon, AC, Natesan, S, Crum, WR, et al. Contrasting effects of haloperidol and lithium on rodent brain structure: A magnetic resonance imaging study with postmortem confirmation. Biol Psychiatry. 2012 May 15; 71(10): 855863. DOI 10.1016/j.biopsych.2011.12.004. Epub 2012 January 15. PubMed PMID: 22244831.Google Scholar
Shim, SS, Hammonds, MD, Ganocy, SJ, Calabrese, JR. Effects of sub-chronic lithium treatment on synaptic plasticity in the dentate gyrus of rat hippocampal slices. Prog Neuropsychopharmacol Biol Psychiatry. 2007 March 30; 31(2): 343347.Epub 2006 November 9. PubMed PMID: 17097205.CrossRefGoogle ScholarPubMed
Pan, W, Banks, WA, Fasold, MB, Bluth, J, Kastin, AJ. Transport of brain-derived neurotrophic factor across the blood-brain barrier. Neuropharmacology. 1998 December; 37(12): 15531561. PubMed PMID: 9886678.Google Scholar
Pillai, A, Kale, A, Joshi, S, et al. Decreased BDNF levels in CSF of drug-naive first-episode psychotic subjects: correlation with plasma BDNF and psychopathology. Int J Neuropsychopharmacol. 2010 May; 13(4): 535539. Epub 2009 November 27. PubMed PMID: 19941699.CrossRefGoogle ScholarPubMed
Eker, C, Kitis, O, Taneli, F, et al. Correlation of serum BDNF levels with hippocampal volumes in first episode, medication-free depressed patients. Eur Arch Psychiatry Clin Neurosci. 2010 October; 260(7): 527533.DOI 10.1007/s00406-010-0110-5. Epub 2010 March 20.Google Scholar
Rizos, E, Papathanasiou, M, Michalopoulou, P, et al. Association of serum BDNF levels with hippocampal volumes in first psychotic episode drug-naïve schizophrenic patients. Schizophr Res. 2011 July; 129(2–3): 201204.Google Scholar
Erickson, KI, Voss, MW, Prakash, RS, et al. Exercise training increases size of hippocampus and improves memory. Proc Natl Acad Sci U S A. 2011 February 15; 108(7): 30173022. DOI 10.1073/pnas.1015950108. Epub 2011 January 31. PubMed PMID: 21282661; PubMed Central PMCID: PMC3041121.Google Scholar
Fernandes, BS, Gama, CS, Ceresér, KM, et al. Brain-derived neurotrophic factor (BDNF) as a state-marker of mood episodes in bipolar disorders: a systematic review and meta-regression analysis. J Psychiatr Res. 2011 August; 45(8): 9951004. Epub 2011 May 6. Review. PubMed PMID: 21550050.CrossRefGoogle ScholarPubMed
Hammonds, MD, Shim, SS. Effects of 4-week treatment with lithium and olanzapine on levels of brain- derived neurotrophic factor (BDNF), B-cell CLL/lymphoma 2 and phosphorylated cyclic adenosine monophosphate response element-binding protein in the sub-regions of the hippocampus. Basic Clin Pharmacol Toxicol. 2009 August; 105(2): 113119. Epub 2009 April 17. PubMed PMID: 19486334.Google Scholar
Jornada, LK, Moretti, M, Valvassori, SS, et al. Effects of mood stabilizers on hippocampus and amygdala BDNF levels in an animal model of mania induced by ouabain. J Psychiatr Res. 2010 un; 44(8): 506510. Epub 2009 December 1. PubMed PMID: 19954800.Google Scholar
de Sousa, R, van de Bilt, M, Diniz, B, et al. Lithium increases plasma brain-derived neurotrophic factor in acute bipolar mania: A preliminary 4-week study. Neurosci Lett. 2011 April 20; 494(1): 5456.Google Scholar
Tramontina, JF, Andreazza, AC, Kauer-Sant’anna, M, et al. Brain-derived neurotrophic factor serum levels before and after treatment for acute mania. Neurosci Lett. 2009 March 13; 452(2):111113. Epub 2009 January 15.CrossRefGoogle ScholarPubMed
Pandey, GN, Rizavi, HS, Dwivedi, Y, Pavuluri, MN. Brain-derived neurotrophic factor gene expression in pediatric bipolar disorder: Effects of treatment and clinical response. J Am Acad Child Adolesc Psychiatry. 2008 September; 47(9): 10771085. PubMed PMID: 18664999.Google Scholar
Frey, BN, Andreazza, AC, Ceresér, KM, et al. Effects of mood stabilizers on hippocampus BDNF levels in an animal model of mania. Life Sci. 2006 June 13; 79(3): 281286. Epub 2006 February 7. PubMed PMID: 16460767.Google Scholar
Chen, G, Rajkowska, G, Du, F, Seraji-Bozorgzad, N., Manji, HK. Enhancement of hippocampal neurogenesis by lithium. J. Neurochem. 2000; 75: 17291734.Google Scholar
Hanson, ND, Nemeroff, CB, Owens, MJ. Lithium, but not fluoxetine or the corticotropin-releasing factor receptor 1 receptor antagonist R121919, increases cell proliferation in the adult dentate gyrus. J Pharmacol Exp Ther. 2011 April; 337(1): 180186. DOI 10.1124/jpet.110.175372. Epub 2011 January 10. PubMed PMID: 21220416; PubMed Central PMCID: PMC3063735.Google Scholar
Shim, SS, Hammonds, MD, Mervis, RF. Four weeks lithium treatment alters neuronal dendrites in the rat hippocampus. Int J Neuropsychopharmacol. 2013 July; 16(6): 13731382. DOI 10.1017/S1461145712001423. Epub 2013 January 18.Google Scholar
Rhindress, K, Ikuta, T, Wellington, R, Malhotra, AK, Szeszko, PR. Delineation of hippocampal subregions using T1-weighted magnetic resonance images at 3 Tesla. Brain Struct Funct. 2015 November; 220(6): 32593272. DOI 10.1007/s00429-014-0854-1. Epub 2014 August 1.Google Scholar
Kafantaris, V, Spritzer, L, Doshi, V, Saito, E, Szeszko, PR. Changes in white matter microstructure predict lithium response in adolescents with bipolar disorder. Bipolar Disord. 2017 November; 19(7): 587594. DOI 10.1111/bdi.12544. Epub 2017 October 9. PubMed PMID: 28992395.Google Scholar
Markham, A, Cameron, I, Bains, R, et al. Brain-derived neurotrophic factor-mediated effects on mitochondrial respiratory coupling and neuroprotection share the same molecular signaling pathways. Eur J Neurosci. 2012 February; 35(3): 366374. PubMed PMID: 22288477.Google Scholar
Rybakowski, JK, Czerski, P, Dmitrzak-Weglarz, M, et al. Clinical and pathogenic aspects of candidate genes for lithium prophylactic efficacy. J Psychopharmacol. 2012 March; 26(3): 368373. Epub 2011 Sep 2.Google Scholar

References

Schildkraut, JJ. The catecholamine hypothesis of affective disorders: A review of supporting evidence. Am J Psychiatry. 1965 November 1; 122(5): 509522.Google Scholar
Ashok, AH, Marques, TAR, Jauhar, S, et al. The dopamine hypothesis of bipolar affective disorder: The state of the art and implications for treatment. Mol Psychiatry. 2017 May; 22(5): 666679.Google Scholar
Beyer, DKE, Freund, N. Animal models for bipolar disorder: From bedside to the cage. Int J Bipolar Disord [Internet]. 2017 October 13:Google Scholar
Mania-like behavior induced by disruption of CLOCK | PNAS [Internet]. [cited 2019 Jul 19]. Available from: www.pnas.org/content/104/15/6406.Google Scholar
Sidor, MM, Spencer, SM, Dzirasa, K, et al. Daytime spikes in dopaminergic activity drive rapid mood-cycling in mice. Mol Psychiatry. 2015 November; 20(11): 14061419.Google Scholar
Tye, KM, Mirzabekov, JJ, Warden, MR, et al. Dopamine neurons modulate neural encoding and expression of depression-related behaviour. Nature. 2013 January 24; 493(7433): 537541.Google Scholar
Young, JW, Cope, ZA, Romoli, B, et al. Mice with reduced DAT levels recreate seasonal-induced switching between states in bipolar disorder. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2018; 43(8):17211731.Google Scholar
Murphy, DL, Brodie, HKH, Goodwin, FK, Bunney, WE. Regular induction of hypomania by L -dopa in “bipolar” manic-depressive patients. Nature. 1971 January; 229(5280): 135.Google Scholar
Vlissides, DN, Gill, D, Castelow, J. Bromocriptine-induced mania? Br Med J. 1978 February 25; 1(6111): 510–510.Google Scholar
Jacobs, D, Silverstone, T. Dextroamphetamine-induced arousal in human subjects as a model for mania. Psychol Med. 1986 May; 16(2): 323329.Google Scholar
McTavish, SF, McPherson, MH, Harmer, CJ, et al. Antidopaminergic effects of dietary tyrosine depletion in healthy subjects and patients with manic illness. Br J Psychiatry J Ment Sci. 2001 October; 179: 356360.CrossRefGoogle ScholarPubMed
Anand, A, Darnell, A, Miller, HL, et al. Effect of catecholamine depletion on lithium-induced long-term remission of bipolar disorder. Biol Psychiatry. 1999 April 15; 45(8): 972978.CrossRefGoogle ScholarPubMed
Delay, J, Deniker, P. 38 cas de psychoses traites par la cure prolong&. et continue de 4560 RP. Ann Med Psychol. 1952; 110: 364396.Google Scholar
Klein, DF, Oaks, G. Importance of psychiatric diagnosis in prediction of clinical drug effects. Arch Gen Psychiatry. 1967 January 1; 16(1): 118126.Google Scholar
Baldessarini, RJ, Tondo, L, Vázquez, GH. Pharmacological treatment of adult bipolar disorder. Mol Psychiatry. 2019 February; 24(2): 198217.Google Scholar
Cipriani, A, Barbui, C, Salanti, G, et al. Comparative efficacy and acceptability of antimanic drugs in acute mania: A multiple-treatments meta-analysis. Lancet Lond Engl. 2011 October 8; 378(9799): 13061315.CrossRefGoogle Scholar
Jauhar, S, Young, AH. Controversies in bipolar disorder; role of second-generation antipsychotic for maintenance therapy. Int J Bipolar Disord. 2019 March 27; 7(1): 10.CrossRefGoogle ScholarPubMed
Cariprazine Treatment of Bipolar Depression: A Randomized Double-Blind Placebo-Controlled Phase 3 Study | American Journal of Psychiatry [Internet]. [cited 2019 Jun 26].Google Scholar
Durgam, S, Earley, W, Lipschitz, A, et al. An 8-week randomized, double-blind, placebo-controlled evaluation of the safety and efficacy of cariprazine in patients with bipolar I depression. Am J Psychiatry. 2016 March 1; 173(3): 271281.Google Scholar
Young, AH, McElroy, SL, Bauer, M, et al. A double-blind, placebo-controlled study of quetiapine and lithium monotherapy in adults in the acute phase of bipolar depression (EMBOLDEN I). J Clin Psychiatry. 2010 February; 71(2): 150–62.Google Scholar
Veselinović, T, Paulzen, M, Gründer, G. Cariprazine, a new, orally active dopamine D2/3 receptor partial agonist for the treatment of schizophrenia, bipolar mania and depression. Expert Rev Neurother. 2013 November; 13(11): 11411159.Google Scholar
Lindström, L, Lindström, E, Nilsson, M, Höistad, M. Maintenance therapy with second generation antipsychotics for bipolar disorder – A systematic review and meta-analysis. J Affect Disord. 2017; 15(213): 138150.Google Scholar
Macfadden, W, Alphs, L, Haskins, JT, et al. A randomized, double-blind, placebo-controlled study of maintenance treatment with adjunctive risperidone long-acting therapy in patients with bipolar I disorder who relapse frequently. Bipolar Disord. 2009 December; 11(8): 827839.Google Scholar
Ketter, TA, Sarma, K, Silva, R, et al. Lurasidone in the long-term treatment of patients with bipolar disorder: A 24-week open-label extension study. Depress Anxiety. 2016 May 1; 33(5): 424434.Google Scholar
Szegedi, A, Durgam, S, Mackle, M, et al. Randomized, double-blind, placebo-controlled trial of asenapine maintenance therapy in adults with an acute manic or mixed episode associated with bipolar I disorder. Am J Psychiatry. 2018 01; 175(1): 7179.Google Scholar
Calabrese, JR, Sanchez, R, Jin, N, et al. Efficacy and safety of aripiprazole once-monthly in the maintenance treatment of bipolar I disorder: A double-blind, placebo-controlled, 52-week randomized withdrawal study. J Clin Psychiatry. 2017; 78(3): 324331.Google Scholar
Quiroz, JA, Yatham, LN, Palumbo, JM, et al. Risperidone long-acting injectable monotherapy in the maintenance treatment of bipolar I disorder. Biol Psychiatry. 2010 July 15; 68(2): 156162.Google Scholar
Vieta, E, Montgomery, S, Sulaiman, AH, et al. A randomized, double-blind, placebo-controlled trial to assess prevention of mood episodes with risperidone long-acting injectable in patients with bipolar I disorder. Eur Neuropsychopharmacol J Eur Coll Neuropsychopharmacol. 2012 November; 22(11): 825835.Google Scholar
Kishi, T, Oya, K, Iwata, N. Long-acting injectable antipsychotics for prevention of relapse in bipolar disorder: A systematic review and meta-analyses of randomized controlled trials. Int J Neuropsychopharmacol [Internet]. 2016 September 21; 19(9): pyw038. DOI:10.1093/ijnp/pyw038.Google Scholar
Paul, Cumming. Imaging Dopamine. Cambridge University Press; 2009.Google Scholar
Paul, Cumming. Imaging Dopamine. Cambridge University Press. 2009.Google Scholar
Wagner, HN, Burns, HD, Dannals, RF, et al. Imaging dopamine receptors in the human brain by positron tomography. Science. 1983 September 23; 221(4617): 12641266.Google Scholar
Nord, M, Farde, L. Antipsychotic occupancy of dopamine receptors in schizophrenia. CNS Neurosci Ther. 2011 April 1; 17(2): 97103.Google Scholar
Piccini, P, Pavese, N, Brooks, DJ. Endogenous dopamine release after pharmacological challenges in Parkinson’s disease. Ann Neurol. 2003 May; 53(5): 647653.Google Scholar
Cropley, VL, Fujita, M, Innis, RB, Nathan, PJ. Molecular imaging of the dopaminergic system and its association with human cognitive function. Biol Psychiatry. 2006 May 15; 59(10): 898907.Google Scholar
Garnett, ES, Firnau, G, Nahmias, C. Dopamine visualized in the basal ganglia of living man. Nature. 1983 September 8; 305(5930): 137138.Google Scholar
Barrio, JR, Huang, SC, Yu, DC, et al. Radiofluorinated L-m-tyrosines: New in-vivo probes for central dopamine biochemistry. J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metab. 1996 July; 16(4): 667678.Google Scholar
DeJesus, OT, Murali, D, Kitchen, R, et al. Evaluation of 3-[18 F]fluoro-alpha-fluoromethyl-p-tyrosine as a tracer for striatal tyrosine hydroxylase activity. Nucl Med Biol. 1994 May; 21(4): 663667.Google Scholar
Innis, R, Baldwin, R, Sybirska, E, et al. Single photon emission computed tomography imaging of monoamine reuptake sites in primate brain with [123I]CIT. Eur J Pharmacol. 1991 August 6; 200(2–3): 369370.Google Scholar
Aquilonius, S-M, Bertröm, K, Eckernäs, S-Å, et al. In vivo evaluation of striatal dopamine reuptake sites using 11 C-nomifensine and positron emission tomography. Acta Neurol Scand. 1987 October 1; 76(4): 283287.Google Scholar
Wong, DF, Yung, B, Dannals, RF, et al. In vivo imaging of baboon and human dopamine transporters by positron emission tomography using [11 C]WIN 35,428. Synapse. 1993 October 1; 15(2): 130142.Google Scholar
Volkow, ND, Ding, YS, Fowler, JS, et al. A new PET ligand for the dopamine transporter: Studies in the human brain. J Nucl Med Off Publ Soc Nucl Med. 1995 December; 36(12): 21622168.Google Scholar
Mozley, PD, Stubbs, JB, Plössl, K, et al. Biodistribution and dosimetry of TRODAT-1: a technetium-99 m tropane for imaging dopamine transporters. J Nucl Med Off Publ Soc Nucl Med. 1998 December; 39(12): 20692076.Google Scholar
Halldin, C, Stone-Elander, S, Farde, L, et al. Preparation of 11 C-labelled SCH 23390 for the in vivo study of dopamine D-1 receptors using positron emission tomography. Int J Rad Appl Instrum [A]. 1986; 37(10): 10391043.Google Scholar
Halldin, C, Foged, C, Chou, YH, et al. Carbon-11-NNC 112: a radioligand for PET examination of striatal and neocortical D1-dopamine receptors. J Nucl Med Off Publ Soc Nucl Med. 1998 December; 39(12): 20612068.Google Scholar
Sit, S-Y, Xie, K, Jacutin-Porte, S, et al. (+)-Dinapsoline: An efficient synthesis and pharmacological profile of a novel dopamine agonist. J Med Chem. 2002 August 1; 45(17): 36603668.Google Scholar
Leysen, JE, Gommeren, W, Laduron, PM. Spiperone: A ligand of choice for neuroleptic receptors. 1. Kinetics and characteristics of in vitro binding. Biochem Pharmacol. 1978 February 1; 27(3): 307316.Google Scholar
Köhler, C, Hall, H, Ogren, SO, Gawell, L. Specific in vitro and in vivo binding of 3 H-raclopride. A potent substituted benzamide drug with high affinity for dopamine D-2 receptors in the rat brain. Biochem Pharmacol. 1985 July 1; 34(13):2251–229.Google Scholar
Kung, HF, Pan, S, Kung, MP, et al. In vitro and in vivo evaluation of [123I]IBZM: A potential CNS D-2 dopamine receptor imaging agent. J Nucl Med Off Publ Soc Nucl Med. 1989 January; 30(1): 8892.Google Scholar
Mukherjee, J, Christian, BT, Dunigan, KA, et al. Brain imaging of 18 F-fallypride in normal volunteers: blood analysis, distribution, test-retest studies, and preliminary assessment of sensitivity to aging effects on dopamine D-2/D-3 receptors. Synap N Y N. 2002 December 1; 46(3): 170–88.Google Scholar
Olsson, H, Halldin, C, Swahn, CG, Farde, L. Quantification of [11 C]FLB 457 binding to extrastriatal dopamine receptors in the human brain. J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metab. 1999 October; 19(10): 11641173.Google Scholar
Seeman, P, Ko, F, Willeit, M, McCormick, P, Ginovart, N. Antiparkinson concentrations of pramipexole and PHNO occupy dopamine D2high and D3high receptors. Synapse. 2005 November 1; 58(2): 122128.Google Scholar
Hwang, DR, Kegeles, LS, Laruelle, M. (-)-N-[(11)C]propyl-norapomorphine: A positron-labeled dopamine agonist for PET imaging of D(2) receptors. Nucl Med Biol. 2000 August; 27(6): 533539.Google Scholar
Finnema, SJ, Seneca, N, Farde, L, et al. A preliminary PET evaluation of the new dopamine D2 receptor agonist [11 C]MNPA in cynomolgus monkey. Nucl Med Biol. 2005 May; 32(4): 353360.Google Scholar
Jauhar, S, Veronese, M, Rogdaki, M, et al. Regulation of dopaminergic function: An [18 F]-DOPA PET apomorphine challenge study in humans. Transl Psychiatry. 2017 February 7; 7(2): e1027.Google Scholar
Narendran, R, Frankle, WG, Mason, NS, et al. Positron emission tomography imaging of amphetamine-induced dopamine release in the human cortex: A comparative evaluation of the high affinity dopamine D2/3 radiotracers [11C]FLB 457 and [11C]fallypride. Synap N Y N. 2009 June; 63(6): 447461.Google Scholar
Shotbolt, P, Tziortzi, AC, Searle, GE, et al. Within-subject comparison of [11C]-(+)-PHNO and [11C]raclopride sensitivity to acute amphetamine challenge in healthy humans. J Cereb Blood Flow Metab. 2012 January; 32(1): 127136.Google Scholar
Cropley, VL, Fujita, M, Bara-Jimenez, W, et al. Pre- and post-synaptic dopamine imaging and its relation with frontostriatal cognitive function in Parkinson disease: PET studies with [11C]NNC 112 and [18F]FDOPA. Psychiatry Res. 2008 July 15; 163(2): 171–82.Google Scholar
Hernaus, D, Mehta, MA. Prefrontal cortex dopamine release measured in vivo with positron emission tomography: Implications for the stimulant paradigm. NeuroImage. 2016 November 15; 142: 663667.Google Scholar
Laruelle, Marc. Measuring dopamine synaptic transmission with molecular imaging and pharmacological challenges: The state of the art. In: Gründer, Gerhard, editors. Molecular Imaging in the Clinical Neurosciences. New York: Springer; 2012, pp. 163203.Google Scholar
Zubieta, J-K, Huguelet, P, Ohl, LE, et al. High vesicular monoamine transporter binding in asymptomatic bipolar I disorder: Sex differences and cognitive correlates. Am J Psychiatry. 2000 October 1; 157(10): 16191628.Google Scholar
Yatham, LN, Liddle, PF, Shiah, I-S, et al. PET study of [(18)F]6-fluoro-L-dopa uptake in neuroleptic- and mood-stabilizer-naive first-episode nonpsychotic mania: Effects of treatment with divalproex sodium. Am J Psychiatry. 2002 May; 159(5): 768774.Google Scholar
Jauhar, S, Nour, MM, Veronese, M, et al. A test of the transdiagnostic dopamine hypothesis of psychosis using positron emission tomographic imaging in bipolar affective disorder and schizophrenia. JAMA Psychiatry. 2017 October 11; 74(12): 12061213.Google Scholar
Anand, A, Verhoeff, P, Seneca, N, et al. Brain SPECT imaging of amphetamine-induced dopamine release in euthymic bipolar disorder patients. Am J Psychiatry. 2000 July; 157(7): 11081114.Google Scholar
Anand, A, Barkay, G, Dzemidzic, M, et al. Striatal dopamine transporter availability in unmedicated bipolar disorder. Bipolar Disord. 2011; 13(4): 406413.Google Scholar
Amsterdam, JD, Newberg, AB. A preliminary study of dopamine transporter binding in bipolar and unipolar depressed patients and healthy controls. Neuropsychobiology. 2007; 55(3–4): 167170.Google Scholar
Chang, TT, Yeh, TL, Chiu, NT, et al. Higher striatal dopamine transporters in euthymic patients with bipolar disorder: A SPECT study with [99mTc] TRODAT-1. Bipolar Disord. 2010 February 1; 12(1): 102106.Google Scholar
Yatham, LN, Liddle, PF, Lam, RW, et al. PET study of the effects of valproate on dopamine D2 receptors in neuroleptic- and mood-stabilizer-naive patients with nonpsychotic mania. Am J Psychiatry. 2002 October 1; 159(10): 17181723.Google Scholar
Suhara, T, Nakayama, K, Inoue, O, et al. D1 dopamine receptor binding in mood disorders measured by positron emission tomography. Psychopharmacology (Berl). 1992; 106(1): 1418.Google Scholar
Pearlson, GD, Wong, DF, Tune, LE, et al. In vivo D2 dopamine receptor density in psychotic and nonpsychotic patients with bipolar disorder. Arch Gen Psychiatry. 1995 June; 52(6): 471477.Google Scholar
Leung, K. 3-N-[11C]methylspiperone. In: Molecular Imaging and Contrast Agent Database (MICAD) [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2004 [cited 2019 July 20].Google Scholar
Jauhar, S, Howes, OD. Understanding and predicting variability in response to treatment in psychotic disorders: In vivo findings. Clin Pharmacol Ther. 2019 May; 105(5): 10791081.Google Scholar
Jauhar, S, Veronese, M, Nour, MM, et al. Determinants of treatment response in first-episode psychosis: An 18 F-DOPA PET study. Mol Psychiatry. 2019 October; 24(10): 15021512.Google Scholar
Jauhar, S, Ratheesh, A, Davey, C, et al. The case for improved care and provision of treatment for people with first-episode mania. Lancet Psychiatry [Internet]. 2019 October 01; 6(10): P869876.Google Scholar
Egerton, A, Demjaha, A, McGuire, P, Mehta, MA, Howes, OD. The test-retest reliability of 18F-DOPA PET in assessing striatal and extrastriatal presynaptic dopaminergic function. Neuroimage. 2010 Apr 1;50(2):524–31.Google Scholar

References

Cipriani, A, Furukawa, TA, Salanti, G, et al. Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: a systematic review and network meta-analysis. Lancet 2018; 391: 13571366.Google Scholar
Warden, D, Rush, AJ, Trivedi, MH, et al. The STAR*D Project results: a comprehensive review of findings. Curr Psychiatry Rep 2007; 9: 449459.Google Scholar
Ferrari, AJ, Charlson, FJ, Norman, RE, et al. Burden of depressive disorders by country, sex, age,and year: findings from the global burden of disease study. PLoS Med 2013; 10: e1001547.Google Scholar
Mayberg, HS. Limbic-cortical dysregulation: a proposed model of depression. Journal of Neuropsychiatry & Clinical Neurosciences 1997; 9: 471481.Google Scholar
Drevets, WC. Neuroimaging and neuropathological studies of depression: implications for the cognitive–emotional features of mood disorders. Current Opinion in Neurobiology 2001; 11: 240249.Google Scholar
Mayberg, HS. Modulating dysfunctional limbic-cortical circuits in depression: towards development of brain-based algorithms for diagnosis and optimised treatment. British Medical Bulletin 2003; 65: 193207.Google Scholar
Roiser, JP, Elliott, R, Sahakian, BJ. Cognitive mechanisms of treatment in depression. Neuropsychopharmacology 2012; 37: 117136.Google Scholar
Peters, SK, Dunlop, K, Downar, J. Cortico-striatal-thalamic loop circuits of the salience network: a central pathway in psychiatric disease and treatment. Front Syst Neurosci 2016; 27: 104.Google Scholar
Fettes, P, Schulze, L, Downar, J. Cortico-striatal-thalamic loop circuits of the orbitofrontal cortex: promising therapeutic targets in psychiatric illness. Front Syst Neurosci 2017; 27: 25.Google Scholar
Brakowski, J, Spinelli, S, Dörig, N, et al. Resting state brain network function in major depression – Depression symptomatology, antidepressant treatment effects, future research. J Psychiatr Res 2017; 92: 147159.Google Scholar
Whitfield-Gabrieli, S, Ford, JM. Default mode network activity and connectivity in psychopathology. Annu Rev Clin Psychol 2015; 8: 4976.Google Scholar
Wang, X, Öngür, D, Auerbach, RP, et al. Cognitive vulnerability to major depression: view from the intrinsic network and cross-network Interactions. Harv Rev Psychiatry 2016; 24: 188201.Google Scholar
Graham, J, Salimi-Khorshidi, G, Hagan, C, et al. Meta-analytic evidence for neuroimaging models of depression: state or trait? J Affect Disord 2013; 151: 423431.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. Biol Psychiatry 2001; 50: 651658.Google Scholar
Anand, A, Li, Y, Wang, Y, et al. Antidepressant effect on connectivity of the mood-regulating circuit: an fMRI study. Neuropsychopharmacology 2005; 30: 13341344.Google Scholar
Anand, A, Li, Y, Wang, Y, et al. Reciprocal effects of antidepressant treatment on activity and connectivity of the mood regulating circuit: an fMRI study. J Neuropsychiatry Clin Neurosci 2007; 19: 274282.Google Scholar
Fales, CL, Barch, DM, Rundle, MM, et al. Antidepressant treatment normalizes hypoactivity in dorsolateral prefrontal cortex during emotional interference processing in major depression. J Affect Disord 2009; 112: 206211.Google Scholar
Victor, TA, Furey, ML, Fromm, SJ, et al. Changes in the neural correlates of implicit emotional face processing during antidepressant treatment in major depressive disorder. Int J Neuropsychopharmacol 2013; 16: 21952208.Google Scholar
Williams, LM, Korgaonkar, MS, Song, YC, et al. Amygdala reactivity to emotional faces in the prediction of general and medication-specific responses to antidepressant treatment in the randomized iSPOT-D trial. Neuropsychopharmacology 2015; 40: 23982408.Google Scholar
Fu, CH, Williams, SC, Cleare, AJ, et al. Attenuation of the neural response to sad faces in major depression by antidepressant treatment: a prospective, event-related functional magnetic resonance imaging study. Arch Gen Psychiatry 2004; 61: 877889.Google Scholar
Fu, CH, Williams, SC, Brammer, MJ, et al. Neural responses to happy facial expressions in major depression following antidepressant treatment. Am J Psychiatry 2007; 164: 599607.Google Scholar
Walsh, ND, Williams, SC, Brammer, MJ, et al. A longitudinal functional magnetic resonance imaging study of verbal working memory in depression after antidepressant therapy. Biol Psychiatry 2007; 62: 12361243.Google Scholar
Chen, CH, Suckling, J, Ooi, C, et al. Functional coupling of the amygdala in depressed patients treated with antidepressant medication. Neuropsychopharmacology 2008; 33: 19091918.Google Scholar
Wang, Y, Xu, C, Cao, X, et al. Effects of an antidepressant on neural correlates of emotional processing in patients with major depression. Neurosci Lett 2012; 527: 5559.Google Scholar
Tao, R, Calley, CS, Hart, J, et al. Brain activity in adolescent major depressive disorder before and after fluoxetine treatment. Am J Psychiatr 2012; 169: 381388.Google Scholar
Heller, AS, Johnstone, T, Light, SN, et al. Relationships between changes in sustained fronto-striatal connectivity and positive affect in major depression resulting from antidepressant treatment. Am J Psychiatr 2013; 170: 197206.Google Scholar
Wagner, G, Koch, K, Schachtzabel, C, et al. Differential effects of serotonergic and noradrenergic antidepressants on brain activity during a cognitive control task and neurofunctional prediction of treatment outcome in patients with depression. J Psychiat Neurosci 2010; 35: 247257.Google Scholar
Arnone, D, McKie, S, Elliott, R, et al. Increased amygdala responses to sad but not fearful faces in major depression: relation to mood state and pharmacological treatment. Am J Psychiatr 2012, 169: 841850.Google Scholar
Jiang, W, Yin, Z, Pang, Y, et al. Brain functional changes in facial expression recognition in patients with major depressive disorder before and after antidepressant treatment: A functional magnetic resonance imaging study. Neural Regen Res 2012; 7: 11511157.Google Scholar
Stoy, M, Schlagenhauf, F, Sterzer, P, et al. Hyporeactivity of ventral striatum towards incentive stimuli in unmedicated depressed patients normalizes after treatment with escitalopram. J Psychopharmacol 2012; 26: 677688.Google Scholar
Godlewska, BR, Norbury, R, Cowen, PJ, et al. Short-term SSRI treatment normalizes amygdala hyperactivity in depressed patients. Psych Medicine 2012; 42: 26092617.Google Scholar
Rosenblau, G, Sterzer, P, Stoy, M, et al. Functional neuroanatomy of emotion processing in major depressive disorder is altered after successful antidepressant therapy. J Psychopharmacol 2012; 26: 14241433.Google Scholar
Miller, JM, Schneck, N, Siegle, GJ, et al. fMRI response to negative words and SSRI treatment outcome in major depressive disorder: a preliminary study. Psychiat Res 2013, 214: 296305.Google Scholar
Wang, L, Li, K, Zhang, Q, et al. Short-term effects of escitalopram on regional brain function in first-episode drug-naive patients with major depressive disorder assessed by resting-state functional magnetic resonance imaging. Psychol Med 2014; 44: 14171426.Google Scholar
Ruhe, HG, Booij, J, Veltman, DJ, et al. Successful pharmacologic treatment of major depressive disorder attenuates amygdala activation to negative facial expressions: a functional magnetic resonance imaging study. J Clin Psychiat 2012; 73: 451459.Google Scholar
Davidson, RJ, Irwin, W, Anderle, MJ, et al. The neural substrates of affective processing in depressed patients treated with venlafaxine. Am J Psychiatr 2003, 160: 6475.Google Scholar
Schaefer, KM Putnam, HS, Benca, RM, et al. Event related functional magnetic resonance imaging measures of neural activity to positive social stimuli in pre- and post-treatment depression. Biol Psychiat 2006, 60: 974986.Google Scholar
Benedetti, F, Radaelli, D, Bernasconi, A, et al. Changes in medial prefrontal cortex neural responses parallel successful antidepressant combination of venlafaxine and light therapy. Arch Ital Biol 2009; 147: 8393.Google Scholar
Lisiecka, D, Meisenzahl, E, Scheuerecker, J, et al. Neural correlates of treatment outcome in major depression. Int J Neuropsychoph 2011; 14: 521534.Google Scholar
Samson, AC, Meisenzahl, E, Scheuerecker, J, et al. Brain activation predicts treatment improvement in patients with major depressive disorder. J Psychiat Res 2011; 45: 12141222.Google Scholar
Gyurak, A, Patenaude, B, Korgaonkar, MS, et al. Frontoparietal activation during response inhibition predicts remission to antidepressants in patients with major depression. Biol Psychiatry 2016; 79: 274281.Google Scholar
Rzepa, E, McCabe, C. Anhedonia and depression severity dissociated by dmPFC resting-state functional connectivity in adolescents. J Psychopharmacol 2018; 32: 10671074.Google Scholar
Harmer, CJ, Bhagwagar, Z, Perrett, DI, et al. Acute SSRI administration affects the processing of social cues in healthy volunteers. Neuropsychopharmacology 2003; 28: 148152.Google Scholar
Scheidegger, M, Henning, A, Walter, M, et al. Effects of ketamine on cognition-emotion interaction in the brain. Neuroimage 2016; 124: 815.Google Scholar
Selvaraj, S, Walker, C, Arnone, D, et al. Effect of citalopram on emotion processing in humans: a combined 5-HT1A [11 C]CUMI-101 PET and functional MRI study. Neuropsychopharmacology 2018; 43: 655664.Google Scholar
Ma, Y. Neuropsychological mechanism underlying antidepressant effect: a systematic metaanalysis. Mol Psychiatry 2015; 20: 311–19.Google Scholar
Wessa, M, Lois, G. Brain functional effects of psychopharmacological treatment in major depression: a focus on neural circuitry of affective processing. Curr Neuropharmacol 2015; 13: 466479.Google Scholar
Fu, CH, Steiner, H, Costafreda, SG. Predictive neural biomarkers of clinical response in depression: a meta-analysis of functional and structural neuroimaging studies of pharmacological and psychological therapies. Neurobiol Dis 2013; 52: 7583.Google Scholar
Etkin, A, Egner, T,. Kalisch, R. Emotional processing in anterior cingulate and medial prefrontal cortex. Trends Cogn Sci 2011; 15: 8593.Google Scholar
Pizzagalli, DA. Frontocingulate dysfunction in depression: toward biomarkers of treatment response. Neuropsychopharmacology 2011; 36: 183206.Google Scholar
Arnone, D. Functional MRI findings, pharmacological treatment in major depression and clinical response. Prog Neuropsychopharmacol Biol Psychiatry 2019; 91: 2837.Google Scholar
Downey, D, Dutta, A, McKie, S, et al. Comparing the actions of lanicemine and ketamine in depression: key role of the anterior cingulate. Eur Neuropsychopharmacol. 2016; 26: 9941003.Google Scholar
McGrath, CL, Kelley, ME, Holtzheimer, PE III, et al. Toward a neuroimaging treatment selection biomarker for major depressive disorder. JAMA Psychiatry 2013; 70: 821829.Google Scholar
Keedwell, P, Drapier, D, Surguladze, S, et al. Neural markers of symptomatic improvement during antidepressant therapy in severe depression: subgenual cingulate and visual cortical responses to sad, but not happy, facial stimuli are correlated with changes in symptom score. J Psychopharmacol 2009; 23: 775788.Google Scholar
Boku, S, Nakagawa, S, Toda, H, et al. Neural basis of major depressive disorder: Beyond monoamine hypothesis. Psychiatry Clin Neurosci 2018; 72: 312.CrossRefGoogle ScholarPubMed
Dusi, N, Barlati, S, Vita, A, et al. Brain structural effects of antidepressant treatment in major depression. Curr Neuropharmacol 2015; 13: 458465.Google Scholar
Gunning, FM, Cheng, J, Murphy, CF, et al. Anterior cingulate cortical volumes and treatment remission of geriatric depression. Int J Geriatr Psychiatry 2009; 24: 829836.Google Scholar
Korgaonkar, MS, Williams, LM, Song, YJ, et al. Diffusion tensor imaging predictors of treatment outcomes in major depressive disorder. Br J Psychiatry 2014; 205: 321328.Google Scholar
Korgaonkar, MS, Rekshan, W, Gordon, E, et al. Magnetic resonance imaging measures of brain structure to predict antidepressant treatment outcome in major depressive disorder. E Bio Medicine 2014; 2: 3745.Google Scholar
Dichter, GS, Gibbs, D, Smoski, MJ. A systematic review of relations between resting-state functional-MRI and treatment response in major depressive disorder. J Affect Disord 2015; 172: 817.Google Scholar
Yang, R, Zhang, H, Wu, X, et al. Hypothalamus-anchored resting brain network changes before and after sertraline treatment in major depression. Biomed Res Int 2014; 2014: 915026.Google Scholar
Posner, J, Hellerstein, DJ, Gat, I, et al. Antidepressants normalize the default mode network in patients with dysthymia. JAMA Psychiatry 2013; 70: 373382.Google Scholar
Li, B, Liu, L, Friston, KJ, et al. A treatment-resistant default mode subnetwork in major depression. Biol Psychiatry 2013; 74: 4854.Google Scholar
Wagner, G, de la Cruz, F, Köhler, S, et al. Treatment associated changes of functional connectivity of midbrain/brainstem nuclei in major depressive disorder. Sci Rep 2017; 7: 8675.Google Scholar
de Kwaasteniet, B, Ruhe, E, Caan, M, et al. Relation between structural and functional connectivity in major depressive disorder. Biol Psychiatry 2013; 74: 4047.Google Scholar
Guo, WB, Liu, F, Chen, JD, et al. Abnormal neural activity of brain regions in treatment-resistant and treatment-sensitive major depressive disorder: a resting-state fMRI study. J Psychiatr Res 2012; 46: 13661373.Google Scholar
Guo, W, Liu, F, Xue, Z, et al. Abnormal resting-state cerebellar-cerebral functional connectivity in treatment-resistant depression and treatment sensitive depression. Prog Neuropsychopharmacol Biol Psychiatry 2013; 44: 5157.Google Scholar
Maltbie, EA, Kaundinya, GS, Howell, LL. Ketamine and pharmacological imaging: use of functional magnetic resonance imaging to evaluate mechanisms of action. Behav Pharmacol 2017; 28: 610622.Google Scholar
Abdallah, CG, Averill, LA, Collins, KA, et al. Ketamine treatment and global brain connectivity in major depression. Neuropsychopharmacology 2017; 42:12101219.Google Scholar
Gopinath, K, Maltbie, E, Urushino, N, et al. Ketamine-induced changes in connectivity of functional brain networks in awake female nonhuman primates: a translational functional imaging model. Psychopharmacology (Berl) 2016; 233: 36733684.Google Scholar
da Cunha-Bang, S, Ettrup, A, Mc Mahon, B, et al. Measuring endogenous changes in serotonergic neurotransmission with [11 C]Cimbi-36 positron emission tomography in humans. Transl Psychiatry 2019; 9: 134.Google Scholar
Miller, JM, Hesselgrave, N, Ogden, RT, et al. Brain serotonin 1 A receptor binding as a predictor of treatment outcome in major depressive disorder. Biol Psychiatry 2013, 15; 74: 760767.Google Scholar
Gray, NA, Milak, MS, DeLorenzo, C, et al. Antidepressant treatment reduces serotonin-1A autoreceptor binding in major depressive disorder. Biol Psychiatry 2013; 74: 2631.Google Scholar
Spies, M, Knudsen, GM, Lanzenberger, R, et al. The serotonin transporter in psychiatric disorders: insights from PET imaging. Lancet Psychiatry 2015; 2: 743755.Google Scholar
Miller, JM, Oquendo, MA, Ogden, RT, et al. Serotonin transporter binding as a possible predictor of one-year remission in major depressive disorder. J Psychiatr Res 2008; 42: 11371144.Google Scholar
Lanzenberger, R, Kranz, GS, Haeusler, D, et al. Prediction of SSRI treatment response in major depression based on serotonin transporter interplay between median raphe nucleus and projection areas. Neuroimage 2012; 63: 874–871.Google Scholar
Yeh, YW, Ho, PS, Kuo, SC, et al. Disproportionate reduction of serotonin transporter may predict the response and adherence to antidepressants in patients with major depressive disorder: a positron emission tomography study with 4-[18 F]-ADAM. Int J Neuropsychopharmacol. 2015 Jan 7;18(7):pyu120.Google Scholar
James, GM, Baldinger-Melich, P, Philippe, C, et al. Effects of selective serotonin reuptake inhibitors on interregional relation of serotonin transporter availability in major depression. Front Hum Neurosci 2017; 11: 48.Google Scholar
Arakawa, R, Stenkrona, P, Takano, A, et al. Venlafaxine ER blocks the norepinephrine transporter in the brain of patients with major depressive disorder: a PET study using [18 F]FMeNER-D2. Int J Neuropsychopharmacol 2019; 22: 278285.Google Scholar
Moriguchi, S, Takano, H, Kimura, Y, et al. Occupancy of norepinephrine transporter by duloxetine in human brains measured by positron emission tomography with (S,S)-[18 F]FMeNER-D2. Int J Neuropsychopharmacol 2017; 20: 957962.Google Scholar
Pringle, A, Harmer, CJ. The effects of drugs on human models of emotional processing: an account of antidepressant drug treatment. Dialogues Clin Neurosci 2015; 17: 477487.Google Scholar
Godlewska, BR. Cognitive neuropsychological theory: reconciliation of psychological and biological approaches for depression. Pharmacol Ther 2018; pii: S0163–7258(18)3023230238.Google Scholar
Komulainen, E, Heikkilä, R, Nummenmaa, L, et al. Short-term escitalopram treatment normalizes aberrant self-referential processing in major depressive disorder. J Affect Disord 2018; 236: 222229.Google Scholar
Godlewska, BR, Browning, M, Norbury, R, et al. Early changes in emotional processing as a marker of clinical response to SSRI treatment in depression. Transl Psych 2016; 6: e957.Google Scholar
Shiroma, PR, Thuras, P, Johns, B, et al. Emotion recognition processing as early predictor of response to 8-week citalopram treatment in late-life depression. Int J Geriatr Psychiatry 2014; 29: 11321139.Google Scholar
MacLeod, C, Rutherford, E, Campbell, L, et al. Selective attention and emotional vulnerability: assessing the causal basis of their association through the experimental manipulation of attentional bias. J Abnorm Psychol 2002; 111: 107123.Google Scholar
Miller, S, McTeague, LM, Gyurak, A, et al. Cognition-childhood maltreatment interactions in the prediction of antidepressant outcomes in major depressive disorder patients: results from the iSPOT-D trial. Depress Anxiety 2015; 32: 594604.Google Scholar
Holmes, EA, Ghaderi, A, Harmer, CJ, et al. Commission on psychological treatments research in tomorrow’s science. The Lancet Psychiatry 2018; 5: 237286.Google Scholar
Browning, M, Reid, C, Cowen, PJ, Goodwin, GM, Harmer, CJ. A single dose of citalopram increases fear recognition in healthy subjects. J Psychopharmacol 2007; 21: 684690.Google Scholar
Jonassen, R, Chelnokova, O, Harmer, C, et al. A single dose of antidepressant alters eye-gaze patterns across face stimuli in healthy women. Psychopharmacology 2014; 232: 953958.Google Scholar
Di Simplicio, M, Norbury, R, Reinecke, A, et al. Paradoxical effects of short-term antidepressant treatment in fMRI emotional processing models in volunteers with high neuroticism. Psychol Med 2014; 44: 241252.Google Scholar
McArthur, RA. Aligning physiology with psychology: translational neuroscience in neuropsychiatric drug discovery. Neuroscience & Biobehavioral Reviews 2017; 76: 421.Google Scholar
Insel, TR. The NIMH Research Domain Criteria (RDoC) Project: precision medicine for psychiatry. Am J Psychiatry 2014; 171: 395397.Google Scholar
Drysdale, AT, Grosenick, L, Downar, J, et al. Resting-state Connectivity Biomarkers Define Neurophysiological Subtypes of Depression Nat Med. 2017 Jan;23(1):28–38.Google Scholar

References

Whiteford, HA, et al. Global burden of disease attributable to mental and substance use disorders: Findings from the global burden of disease study 2010. Lancet. 2013; 382(9904): 15751586.Google Scholar
Park, SC, et al. Does age at onset of first major depressive episode indicate the subtype of major depressive disorder?: The clinical research center for depression study. Yonsei Med J. 2014; 55(6): 17121720.Google Scholar
Kessler, RC, et al. Age differences in the prevalence and co-morbidity of DSM-IV major depressive episodes: Results from the WHO world mental health survey initiative. Depress Anxiety. 2010; 27(4): 351364.Google Scholar
Oquendo, MA. McGrath, P, Weissman, MM. Biomarker studies and the future of personalized treatment for depression. Depression and Anxiety. 2014; 31(11): 902905.Google Scholar
Chen, C-H, et al. Brain imaging correlates of depressive symptom severity and predictors of symptom improvement after antidepressant treatment. Biological Psychiatry. 2007; 62(5): 407414.Google Scholar
Li, C-T, et al. Cognition-modulated frontal activity in prediction and augmentation of antidepressant efficacy: A randomized controlled pilot study. Cerebral Cortex. 2014; 26(1): 202210.Google Scholar
Cuijpers, P, et al. The effects of psychotherapies for major depression in adults on remission, recovery and improvement: A meta-analysis. Journal of Affective Disorders. 2014; 159: 118126.Google Scholar
Cuijpers, P, et al. Psychological treatment of depression: A meta-analytic database of randomized studies. BMC Psychiatry. 2008; 8(1): 36.Google Scholar
Cuijpers, P, et al. A meta-analysis of cognitive-behavioural therapy for adult depression, alone and in comparison with other treatments. The Canadian Journal of Psychiatry. 2013; 58(7): 376385.Google Scholar
Karyotaki, E, et al. Combining pharmacotherapy and psychotherapy or monotherapy for major depression? A meta-analysis on the long-term effects. Journal of Affective Disorders. 2016; 194: 144152.Google Scholar
Cuijpers, P, et al. Adding psychotherapy to antidepressant medication in depression and anxiety disorders: A meta‐analysis. World Psychiatry. 2014; 13(1): 5667.Google Scholar
Chakrabarty, T, Ogrodniczuk, J, Hadjipavlou, G. Predictive neuroimaging markers of psychotherapy response: A systematic review. Harvard Review of Psychiatry. 2016; 24(6): 396405.Google Scholar
Linden, D. How psychotherapy changes the brain–the contribution of functional neuroimaging. Molecular Psychiatry, 2006. 11(6): 528.Google Scholar
Frewen, PA, Dozois, DJ, Lanius, RA. Neuroimaging studies of psychological interventions for mood and anxiety disorders: empirical and methodological review. Focus. 2010; 8(1): 92109.Google Scholar
Beauregard, M. Functional neuroimaging studies of the effects of psychotherapy. Dialogues in Clinical Neuroscience. 2014; 16(1): 75.Google Scholar
Barsaglini, A, et al. The effects of psychotherapy on brain function: A systematic and critical review. Progress in Neurobiology. 2014; 114: 114.Google Scholar
Roffman, JL, et al. Neuroimaging and the functional neuroanatomy of psychotherapy. Psychological Medicine. 2005; 35(10): 13851398.Google Scholar
Fu, C, Steiner, H, Costafreda, SG. Predictive neural biomarkers of clinical response in depression: a meta-analysis of functional and structural neuroimaging studies of pharmacological and psychological therapies. Neurobiol Disease. 2013. 52: 7583.Google Scholar
Beck, JS. Cognitive Behavior Therapy: Basics and Beyond. Guilford press; 2011.Google Scholar
Parker, G, Fletcher, K. Treating depression with the evidence‐based psychotherapies: A critique of the evidence. Acta Psychiatrica Scandinavica. 2007; 115(5): 352359.Google Scholar
Beck, AT. Cognitive Therapy of Depression. Guilford press; 1979.Google Scholar
Crowther, A, et al. Resting-state connectivity predictors of response to psychotherapy in major depressive disorder. Neuropsychopharmacology. 2015; 40(7): 16591673.Google Scholar
Dimidjian, S, et al. Randomized trial of behavioral activation, cognitive therapy, and antidepressant medication in the acute treatment of adults with major depression. Journal of Consulting and Clinical Psychology. 2006; 74(4): 658.Google Scholar
Dobson, KS, et al. Randomized trial of behavioral activation, cognitive therapy, and antidepressant medication in the prevention of relapse and recurrence in major depression. Journal of Consulting and Clinical Psychology. 2008; 76(3): 468.Google Scholar
Klerman, GL, et al. Interpersonal Psychotherapy for Depression. Lanham, Maryland, USA: Rowman & Littlefield; 1996.Google Scholar
Cuijpers, P, et al. Interpersonal psychotherapy for depression: A meta-analysis. American Journal of Psychiatry. 2011; 168(6): 581592.Google Scholar
Weingarten, CP, Strauman, TJ. Neuroimaging for psychotherapy research: current trends. Psychotherapy Research. 2015; 25(2): 185213.Google Scholar
Fournier, JC, Price, RB. Psychotherapy and neuroimaging. Focus. 2014; 12(3): 290298.Google Scholar
Drevets, WC. Neuroimaging and neuropathological studies of depression: Implications for the cognitive-emotional features of mood disorders. Current opinion in neurobiology. 2001; 11(2): 240249.Google Scholar
Hariri, AR, Bookheimer, SY, Mazziotta, JC. Modulating emotional responses: Effects of a neocortical network on the limbic system. Neuroreport. 2000; 11(1): 4348.Google Scholar
Fales, CL, et al. Altered emotional interference processing in affective and cognitive-control brain circuitry in major depression. Biological Psychiatry. 2008; 63(4): 377384.Google Scholar
Vargas, C, Lopez-Jaramillo, C, Vieta, E. A systematic literature review of resting state network–functional MRI in bipolar disorder. J Affect Disord. 2013; 150(3): 727735.Google Scholar
Radaelli, D, et al. Fronto-limbic disconnection in bipolar disorder. Eur Psychiatry. 2015; 30(1): 8288.Google Scholar
Phillips, ML, Swartz, HA. A critical appraisal of neuroimaging studies of bipolar disorder: toward a new conceptualization of underlying neural circuitry and a road map for future research. Am J Psychiatry. 2014; 171(8): 829843.Google Scholar
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
Yoshimura, S, et al. Cognitive behavioral therapy for depression changes medial prefrontal and ventral anterior cingulate cortex activity associated with self-referential processing. Social Cognitive and Affective Neuroscience. 2014; 9(4): 487493.Google Scholar
Ritchey, M, et al. Neural correlates of emotional processing in depression: Changes with cognitive behavioral therapy and predictors of treatment response. Journal of Psychiatric Research. 2011; 45(5): 577587.Google Scholar
Fu, C, et al. Neural responses to sad facial expressions in major depression following cognitive behavioral therapy. Biological Psychiatry. 2008; 64(6): 505512.Google Scholar
Yoshimura, S, et al. Cognitive behavioral therapy changes functional connectivity between medial prefrontal and anterior cingulate cortices. Journal of Affective Disorders 2017; 208: 610614.Google Scholar
Assaf, M, et al. Neural functional architecture and modulation during decision making under uncertainty in individuals with generalized anxiety disorder. Brain Behav. 2018 August; 8(8): e01015.Google Scholar
Kennedy, SH, et al. Differences in brain glucose metabolism between responders to CBT and venlafaxine in a 16-week randomized controlled trial. American Journal of Psychiatry. 2007; 164(5): 778788.Google Scholar
Konarski, JZ, et al. Relationship between regional brain metabolism, illness severity and age in depressed subjects. Psychiatry Research: Neuroimaging. 2007; 155(3): 203210.Google Scholar
Dichter, GS, Felder, JN, Smoski, MJ. The effects of brief behavioral activation therapy for depression on cognitive control in affective contexts: An fMRI investigation. Journal of Affective Disorders 2010; 126(1): 236244.Google Scholar
Burgess, PW, Dumontheil, I, Gilbert, SI. The gateway hypothesis of rostral prefrontal cortex (area 10) function. Trends in Cognitive Sciences. 2007; 11(7): 290298.Google Scholar
Wright, P, et al. Dissociated responses in the amygdala and orbitofrontal cortex to bottom–up and top–down components of emotional evaluation. Neuroimage. 2008; 39(2): 894902.Google Scholar
Jollant, F, et al. Decreased activation of lateral orbitofrontal cortex during risky choices under uncertainty is associated with disadvantageous decision-making and suicidal behavior. Neuroimage 2010; 51(3): 12751281.Google Scholar
Fales, CL, et al. Antidepressant treatment normalizes hypoactivity in dorsolateral prefrontal cortex during emotional interference processing in major depression. Journal of Affective Disorders. 2009; 112(1–3): 206211.Google Scholar
Harmer, CJ, et al. Antidepressant drug treatment modifies the neural processing of nonconscious threat cues. Biological Psychiatry. 2006; 59(9): 816820.Google Scholar
Tiger, M, et al. Reduced 5-HT(1B) receptor binding in the dorsal brain stem after cognitive behavioural therapy of major depressive disorder. Psychiatry Res. 2014; 223(2): 164170.Google Scholar
Goldapple, K, et al. Modulation of cortical-limbic pathways in major depression: Treatment-specific effects of cognitive behavior therapy. Archives of General Psychiatry. 2004; 61(1): 3441.Google Scholar
Brody, AL, et al. Regional brain metabolic changes in patients with major depression treated with either paroxetine or interpersonal therapy: Preliminary findings. Archives of General Psychiatry. 2001; 58(7): 631640.Google Scholar
Martin, SD, et al. Brain blood flow changes in depressed patients treated with interpersonal psychotherapy or venlafaxine hydrochloride: Preliminary findings. Archives of General Psychiatry. 2001; 58(7): 641648.Google Scholar
Sanacora, G, et al. Increased cortical GABA concentrations in depressed patients receiving ECT. American Journal of Psychiatry. 2003; 160(3): 577579.Google Scholar
Sanacora, G, et al. Cortical γ-aminobutyric acid concentrations in depressed patients receiving cognitive behavioral therapy. Biological Psychiatry 2006; 59(3): 284286.Google Scholar
Abdallah, CG, et al. Decreased occipital cortical glutamate levels in response to successful cognitive-behavioral therapy and pharmacotherapy for major depressive disorder. Psychother Psychosom. 2014; 83(5): 298307.Google Scholar
Costafreda, SG, et al. Neural correlates of sad faces predict clinical remission to cognitive behavioural therapy in depression. Neuroreport. 2009; 20(7): 637641.Google Scholar
Siegle, GJ, et al. Toward clinically useful neuroimaging in depression treatment: Prognostic utility of subgenual cingulate activity for determining depression outcome in cognitive therapy across studies, scanners, and patient characteristics. Archives of General Psychiatry. 2012; 69(9): 913924.Google Scholar
Ressler, KJ. Amygdala activity, fear, and anxiety: Modulation by stress. Biological Psychiatry. 2010; 67(12): 11171119.Google Scholar
Dunlop, BW, et al. Functional connectivity of the subcallosal cingulate cortex and differential outcomes to treatment with cognitive-behavioral therapy or antidepressant medication for major depressive disorder. Am J Psychiatry. 2017; 174(6): 533545.Google Scholar
Thompson, DG, et al. fMRI activation during executive function predicts response to cognitive behavioral therapy in older, depressed adults. American Journal of Geriatric Psychiatry 2015; 23(1): 1322.Google Scholar
Forbes, EE, et al. Reward-related brain function as a predictor of treatment response in adolescents with major depressive disorder. Cognitive, Affective, & Behavioral Neuroscience. 2010; 10(1): 107118.Google Scholar
Fu, C, et al. Neural responses to sad facial expressions in major depression following cognitive behavioral therapy Biol Psychiatry. 2008; 64(6): 505512.Google Scholar
Carl, H, et al. Sustained anterior cingulate cortex activation during reward processing predicts response to psychotherapy in major depressive disorder. J Affect Disord. 2016; 203: 204212.Google Scholar
Dichter, GS, Felder, JN, Bodfish, JW. Autism is characterized by dorsal anterior cingulate hyperactivation during social target detection. Social Cognitive and Affective Neuroscience. 2009; 4(3): 215226.Google Scholar
McGrath, CL, et al. Pretreatment brain states identify likely nonresponse to standard treatments for depression. Biological Psychiatry. 2014; 76(7): 527535.Google Scholar
Yoshimura, S, et al. Cognitive behavioral therapy for depression changes medial prefrontal and ventral anterior cingulate cortex activity associated with self-referential processing. Social Cognitive and Affective Neuroscience. 2013; 9(4): 487493.Google Scholar
Konarski, JZ, et al. Predictors of nonresponse to cognitive behavioural therapy or venlafaxine using glucose metabolism in major depressive disorder. Journal of Psychiatry & Neuroscience: JPN. 2009; 34(3): 175.Google Scholar
Seminowicz, D, et al. Limbic–frontal circuitry in major depression: A path modeling metanalysis. Neuroimage. 2004; 22(1): 409418.Google Scholar
Critchley, HD, et al. Neural systems supporting interoceptive awareness. Nature Neuroscience. 2004; 7(2): 189.Google Scholar
Fujino, J, et al. Anterior cingulate volume predicts response to cognitive behavioral therapy in major depressive disorder. J Affect Disord. 2015; 174: 397399.Google Scholar
Sambataro, F, et al. Anterior cingulate volume predicts response to psychotherapy and functional connectivity with the inferior parietal cortex in major depressive disorder. Eur Neuropsychopharmacol. 2018; 28(1): 138148.Google Scholar
Pizzagalli, DA. Frontocingulate dysfunction in depression: Toward biomarkers of treatment response. Neuropsychopharmacology. 2011; 36(1): 183.Google Scholar
Burkhouse, KL, et al. Neural reactivity to reward as a predictor of cognitive behaviroal therapy response in anxiety and depression. Depress Anxiety. 2016; 33(4): 281288.Google Scholar
DeRubeis, RJ, Strunk, DR. The Oxford Handbook of Mood Disorders. Oxford University Press; 2017.Google Scholar
Salvadore, G, 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): 1415.Google Scholar
Keedwell, P, et al. Neural markers of symptomatic improvement during antidepressant therapy in severe depression: Subgenual cingulate and visual cortical responses to sad, but not happy, facial stimuli are correlated with changes in symptom score. Journal of Psychopharmacology 2009; 23(7): 775788.Google Scholar
Weigand, A, et al. Prospective validation that subgenual connectivity predicts antidepressant efficacy of transcranial magnetic stimulation sites. Biological Psychiatry. 2018; 84(1): 2837.Google Scholar
Wu, J, et al. Prediction of antidepressant effects of sleep deprivation by metabolic rates in the ventral anterior cingulate and medial prefrontal cortex. American Journal of Psychiatry. 1999; 156(8): 11491158.Google Scholar
Kringelbach, ML, Rolls, ET. The functional neuroanatomy of the human orbitofrontal cortex: Evidence from neuroimaging and neuropsychology. Progress in Neurobiology. 2004; 72(5): 341372.Google Scholar
Kurth, F, et al. A link between the systems: Functional differentiation and integration within the human insula revealed by meta-analysis. Brain Structure and Function. 2010; 214(5–6): 519534.Google Scholar
van der Werff, SJ, et al. Neuroimaging resilience to stress: A review. Frontiers in Behavioral Neuroscience. 2013; 7: 39.Google Scholar
Drevets, WC, Price, JL, Furey, ML. Brain structural and functional abnormalities in mood disorders: implications for neurocircuitry models of depression. Brain Structure and Function. 2008; 213(1–2): 93118.Google Scholar
Shin, LM, Liberzon, I. The neurocircuitry of fear, stress, and anxiety disorders. Neuropsychopharmacology. 2010; 35(1): 169.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
×