Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Prefrontal cortex circuits in depression and anxiety: contribution of discrete neuronal populations and target regions

Abstract

Our understanding of depression and its treatment has advanced with the advent of ketamine as a rapid-acting antidepressant and the development and refinement of tools capable of selectively altering the activity of populations of neuronal subtypes. This work has resulted in a paradigm shift away from dysregulation of single neurotransmitter systems in depression towards circuit level abnormalities impacting function across multiple brain regions and neurotransmitter systems. Studies on the features of circuit level abnormalities demonstrate structural changes within the prefrontal cortex (PFC) and functional changes in its communication with distal brain structures. Treatments that impact the activity of brain regions, such as transcranial magnetic stimulation or rapid-acting antidepressants like ketamine, appear to reverse depression associated circuit abnormalities though the mechanisms underlying the reversal, as well as development of these abnormalities remains unclear. Recently developed optogenetic and chemogenetic tools that allow high-fidelity control of neuronal activity in preclinical models have begun to elucidate the contributions of the PFC and its circuitry to depression- and anxiety-like behavior. These tools offer unprecedented access to specific circuits and neuronal subpopulations that promise to offer a refined view of the circuit mechanisms surrounding depression and potential mechanistic targets for development and reversal of depression associated circuit abnormalities.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Regional specificity of modalities for transiently manipulating neuronal activity.
Fig. 2: Viral strategies for targeting neuronal populations.
Fig. 3: Preclinical neuronal manipulation methods and mPFC circuitry implicated in depression-like behavior.

Similar content being viewed by others

References

  1. Guilbert JJ. The world health report 2002—reducing risks, promoting healthy life. Educ Health (Abingdon, Engl). 2003;16:230.

    CAS  Google Scholar 

  2. Kessler RC, Berglund P, Demler O, Jin R, Koretz D, Merikangas KR, et al. The epidemiology of major depressive disorder: results from the National Comorbidity Survey Replication (NCS-R). JAMA. 2003;289:3095–105.

    PubMed  Google Scholar 

  3. Greenberg PE, Fournier AA, Sisitsky T, Pike CT, Kessler RC. The economic burden of adults with major depressive disorder in the United States (2005 and 2010). J Clin Psychiatry. 2015;76:155–62.

    PubMed  Google Scholar 

  4. Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, Ritz L, et al. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry. 2006;163:28–40.

    PubMed  Google Scholar 

  5. Flint J, Kendler KS. The genetics of major depression. Neuron. 2014;81:1214.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Border R, Johnson EC, Evans LM, Smolen A, Berley N, Sullivan PF, et al. No support for historical candidate gene or candidate gene-by-interaction hypotheses for major depression across multiple large samples. Am J Psychiatry. 2019;176:appiajp201818070881.

    Google Scholar 

  7. Savitz J, Drevets WC. Bipolar and major depressive disorder: neuroimaging the developmental-degenerative divide. Neurosci Biobehav Rev. 2009;33:699–771.

    PubMed  PubMed Central  Google Scholar 

  8. Duman RS, Sanacora G, Krystal JH. Altered connectivity in depression: GABA and glutamate neurotransmitter deficits and reversal by novel treatments. Neuron. 2019;102:75–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Dubin MJ, Liston C, Avissar MA, Ilieva I, Gunning FM. Network-guided transcranial magnetic stimulation for depression. Curr Behav Neurosci Rep. 2017;4:70–77.

    PubMed  PubMed Central  Google Scholar 

  10. Liston C, Chen AC, Zebley BD, Drysdale AT, Gordon R, Leuchter B, et al. Default mode network mechanisms of transcranial magnetic stimulation in depression. Biol Psychiatry. 2014;76:517–26.

    PubMed  PubMed Central  Google Scholar 

  11. Singh A, Kar SK. How electroconvulsive therapy works?: Understanding the neurobiological mechanisms. Clin Psychopharmacol Neurosci. 2017;15:210–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Mayberg HS, Lozano AM, Voon V, McNeely HE, Seminowicz D, Hamani C, et al. Deep brain stimulation for treatment-resistant depression. Neuron. 2005;45:651–60.

    CAS  PubMed  Google Scholar 

  13. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47:351–4.

    CAS  PubMed  Google Scholar 

  14. Zarate CA Jr., Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63:856–64.

    CAS  PubMed  Google Scholar 

  15. Abdallah CG, De Feyter HM, Averill LA, Jiang L, Averill CL, Chowdhury GMI, et al. The effects of ketamine on prefrontal glutamate neurotransmission in healthy and depressed subjects. Neuropsychopharmacology. 2018;43:2154–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Holmes SE, Scheinost D, Finnema SJ, Naganawa M, Davis MT, DellaGioia N, et al. Lower synaptic density is associated with depression severity and network alterations. Nat Commun. 2019;10:1529.

    PubMed  PubMed Central  Google Scholar 

  17. Kang HJ, Voleti B, Hajszan T, Rajkowska G, Stockmeier CA, Licznerski P, et al. Decreased expression of synapse-related genes and loss of synapses in major depressive disorder. Nat Med. 2012;18:1413–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Voleti B, Navarria A, Liu RJ, Banasr M, Li N, Terwilliger R, et al. Scopolamine rapidly increases mammalian target of rapamycin complex 1 signaling, synaptogenesis, and antidepressant behavioral responses. Biol Psychiatry. 2013;74:742–9.

    CAS  PubMed  Google Scholar 

  19. Li N, Liu RJ, Dwyer JM, Banasr M, Lee B, Son H, et al. Glutamate N-methyl-D-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits caused by chronic stress exposure. Biol Psychiatry. 2011;69:754–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329:959–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiatry. 2006;59:1116–27.

    CAS  PubMed  Google Scholar 

  22. Moda-Sava RN, Murdock MH, Parekh PK, Fetcho RN, Huang BS, Huynh TN, et al. Sustained rescue of prefrontal circuit dysfunction by antidepressant-induced spine formation. Science. 2019;364:147.

  23. Lee AL, Ogle WO, Sapolsky RM. Stress and depression: possible links to neuron death in the hippocampus. Bipolar Disord. 2002;4:117–28.

    CAS  PubMed  Google Scholar 

  24. Fuchs E, Czeh B, Kole MH, Michaelis T, Lucassen PJ. Alterations of neuroplasticity in depression: the hippocampus and beyond. Eur Neuropsychopharmacol. 2004;14:S481–90.

    CAS  PubMed  Google Scholar 

  25. Thompson SM, Kallarackal AJ, Kvarta MD, Van Dyke AM, LeGates TA, Cai X. An excitatory synapse hypothesis of depression. Trends Neurosci. 2015;38:279–94.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Fogaca MV, Duman RS. Cortical GABAergic dysfunction in stress and depression: new insights for therapeutic interventions. Front Cell Neurosci. 2019;13:87.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Moriguchi S, Takamiya A, Noda Y, Horita N, Wada M, Tsugawa S, et al. Glutamatergic neurometabolite levels in major depressive disorder: a systematic review and meta-analysis of proton magnetic resonance spectroscopy studies. Mol Psychiatry. 2018;24:952–64.

  28. Feyissa AM, Chandran A, Stockmeier CA, Karolewicz B. Reduced levels of NR2A and NR2B subunits of NMDA receptor and PSD-95 in the prefrontal cortex in major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33:70–75.

    CAS  PubMed  Google Scholar 

  29. Gray AL, Hyde TM, Deep-Soboslay A, Kleinman JE, Sodhi MS. Sex differences in glutamate receptor gene expression in major depression and suicide. Mol Psychiatry. 2015;20:1057–68.

    CAS  PubMed  Google Scholar 

  30. Karolewicz B, Maciag D, O’Dwyer G, Stockmeier CA, Feyissa AM, Rajkowska G. Reduced level of glutamic acid decarboxylase-67 kDa in the prefrontal cortex in major depression. Int J Neuropsychopharmacol. 2010;13:411–20.

    CAS  PubMed  Google Scholar 

  31. Rajkowska G, O’Dwyer G, Teleki Z, Stockmeier CA, Miguel-Hidalgo JJ. GABAergic neurons immunoreactive for calcium binding proteins are reduced in the prefrontal cortex in major depression. Neuropsychopharmacology. 2007;32:471–82.

    CAS  PubMed  Google Scholar 

  32. Fee C, Banasr M, Sibille E. Somatostatin-positive gamma-aminobutyric acid interneuron deficits in depression: cortical microcircuit and therapeutic perspectives. Biol Psychiatry. 2017;82:549–59.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Bajbouj M, Lisanby SH, Lang UE, Danker-Hopfe H, Heuser I, Neu P. Evidence for impaired cortical inhibition in patients with unipolar major depression. Biol Psychiatry. 2006;59:395–400.

    PubMed  Google Scholar 

  34. Levinson AJ, Fitzgerald PB, Favalli G, Blumberger DM, Daigle M, Daskalakis ZJ. Evidence of cortical inhibitory deficits in major depressive disorder. Biol Psychiatry. 2010;67:458–64.

    CAS  PubMed  Google Scholar 

  35. Sanacora G, Mason GF, Rothman DL, Behar KL, Hyder F, Petroff OA, et al. Reduced cortical gamma-aminobutyric acid levels in depressed patients determined by proton magnetic resonance spectroscopy. Arch Gen Psychiatry. 1999;56:1043–7.

    CAS  PubMed  Google Scholar 

  36. Sanacora G, Gueorguieva R, Epperson CN, Wu YT, Appel M, Rothman DL, et al. Subtype-specific alterations of gamma-aminobutyric acid and glutamate in patients with major depression. Arch Gen Psychiatry. 2004;61:705–13.

    CAS  PubMed  Google Scholar 

  37. Dubin MJ, Mao X, Banerjee S, Goodman Z, Lapidus KA, Kang G, et al. Elevated prefrontal cortex GABA in patients with major depressive disorder after TMS treatment measured with proton magnetic resonance spectroscopy. J Psychiatry Neurosci. 2016;41:E37–45.

    PubMed  PubMed Central  Google Scholar 

  38. Drysdale AT, Grosenick L, Downar J, Dunlop K, Mansouri F, Meng Y, et al. Resting-state connectivity biomarkers define neurophysiological subtypes of depression. Nat Med. 2017;23:28–38.

    CAS  PubMed  Google Scholar 

  39. Lee WH, Lisanby SH, Laine AF, Peterchev AV. Comparison of electric field strength and spatial distribution of electroconvulsive therapy and magnetic seizure therapy in a realistic human head model. Eur Psychiatry. 2016;36:55–64.

    CAS  PubMed  Google Scholar 

  40. Deng ZD, Lisanby SH, Peterchev AV. Electric field strength and focality in electroconvulsive therapy and magnetic seizure therapy: a finite element simulation study. J Neural Eng. 2011;8:016007.

    PubMed  PubMed Central  Google Scholar 

  41. Deng ZD, Lisanby SH, Peterchev AV. Electric field depth-focality tradeoff in transcranial magnetic stimulation: simulation comparison of 50 coil designs. Brain Stimul. 2013;6:1–13.

    PubMed  Google Scholar 

  42. Roth Y, Pell GS, Chistyakov AV, Sinai A, Zangen A, Zaaroor M. Motor cortex activation by H-coil and figure-8 coil at different depths. Combined motor threshold and electric field distribution study. Clin Neurophysiol. 2014;125:336–43.

    PubMed  Google Scholar 

  43. McIntyre CC, Savasta M, Kerkerian-Le Goff L, Vitek JL. Uncovering the mechanism(s) of action of deep brain stimulation: activation, inhibition, or both. Clin Neurophysiol. 2004;115:1239–48.

    PubMed  Google Scholar 

  44. Stujenske JM, Spellman T, Gordon JA. Modeling the spatiotemporal dynamics of light and heat propagation for In Vivo optogenetics. Cell Rep. 2015;12:525–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Watakabe A, Ohtsuka M, Kinoshita M, Takaji M, Isa K, Mizukami H, et al. Comparative analyses of adeno-associated viral vector serotypes 1, 2, 5, 8 and 9 in marmoset, mouse and macaque cerebral cortex. Neurosci Res. 2015;93:144–57.

    PubMed  Google Scholar 

  46. Scheyltjens I, Laramee ME, Van den Haute C, Gijsbers R, Debyser Z, Baekelandt V, et al. Evaluation of the expression pattern of rAAV2/1, 2/5, 2/7, 2/8, and 2/9 serotypes with different promoters in the mouse visual cortex. J Comp Neurol. 2015;523:2019–42.

    CAS  PubMed  Google Scholar 

  47. Hare BD, Shinohara R, Liu RJ, Pothula S, DiLeone RJ, Duman RS. Optogenetic stimulation of medial prefrontal cortex Drd1 neurons produces rapid and long-lasting antidepressant effects. Nat Commun. 2019;10:223.

    PubMed  PubMed Central  Google Scholar 

  48. Nakamura S, Baratta MV, Pomrenze MB, Dolzani SD, Cooper DC. High fidelity optogenetic control of individual prefrontal cortical pyramidal neurons in vivo. F1000Research. 2012;1:7.

    PubMed  PubMed Central  Google Scholar 

  49. Arias-Gil G, Ohl FW, Takagaki K, Lippert MT. Measurement, modeling, and prediction of temperature rise due to optogenetic brain stimulation. Neurophotonics. 2016;3:045007.

    PubMed  PubMed Central  Google Scholar 

  50. Owen SF, Liu MH, Kreitzer AC. Thermal constraints on in vivo optogenetic manipulations. Nat Neurosci. 2019;22:1061–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Tyssowski KM, Gray JM. Blue light increases neuronal activity-regulated gene expression in the absence of optogenetic proteins. eNeuro. 2019;6:1–10.

  52. Alexander GM, Rogan SC, Abbas AI, Armbruster BN, Pei Y, Allen JA, et al. Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptors. Neuron. 2009;63:27–39.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Gomez JL, Bonaventura J, Lesniak W, Mathews WB, Sysa-Shah P, Rodriguez LA, et al. Chemogenetics revealed: DREADD occupancy and activation via converted clozapine. Science. 2017;357:503–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Laubach M, Amarante LM, Swanson K, White SR. What, if anything, is rodent prefrontal cortex? eNeuro. 2018;5:1–14.

  55. Heilbronner SR, Rodriguez-Romaguera J, Quirk GJ, Groenewegen HJ, Haber SN. Circuit-based corticostriatal homologies between rat and primate. Biol Psychiatry. 2016;80:509–21.

    PubMed  PubMed Central  Google Scholar 

  56. Seamans JK, Lapish CC, Durstewitz D. Comparing the prefrontal cortex of rats and primates: insights from electrophysiology. Neurotox Res. 2008;14:249–62.

    PubMed  Google Scholar 

  57. Burgos-Robles A, Vidal-Gonzalez I, Quirk GJ. Sustained conditioned responses in prelimbic prefrontal neurons are correlated with fear expression and extinction failure. J Neurosci. 2009;29:8474–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Courtin J, Chaudun F, Rozeske RR, Karalis N, Gonzalez-Campo C, Wurtz H, et al. Prefrontal parvalbumin interneurons shape neuronal activity to drive fear expression. Nature. 2014;505:92–96.

    PubMed  Google Scholar 

  59. Herry C, Ciocchi S, Senn V, Demmou L, Muller C, Luthi A. Switching on and off fear by distinct neuronal circuits. Nature. 2008;454:600–6.

    CAS  PubMed  Google Scholar 

  60. Fuchikami M, Thomas A, Liu R, Wohleb ES, Land BB, DiLeone RJ, et al. Optogenetic stimulation of infralimbic PFC reproduces ketamine’s rapid and sustained antidepressant actions. Proc Natl Acad Sci USA. 2015;112:8106–11.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Giustino TF, Maren S. The role of the medial prefrontal cortex in the conditioning and extinction of fear. Front Behav Neurosci. 2015;9:298.

    PubMed  PubMed Central  Google Scholar 

  62. Muir J, Lopez J, Bagot RC. Wiring the depressed brain: optogenetic and chemogenetic circuit interrogation in animal models of depression. Neuropsychopharmacology. 2018;44:1013–26.

    PubMed  PubMed Central  Google Scholar 

  63. de Kloet ER, Molendijk ML. Coping with the forced swim stressor: towards understanding an adaptive mechanism. Neural Plasticity. 2016;2016:6503162.

    PubMed  PubMed Central  Google Scholar 

  64. Warden MR, Selimbeyoglu A, Mirzabekov JJ, Lo M, Thompson KR, Kim SY, et al. A prefrontal cortex-brainstem neuronal projection that controls response to behavioural challenge. Nature. 2012;492:428–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Kumar S, Black SJ, Hultman R, Szabo ST, DeMaio KD, Du J, et al. Cortical control of affective networks. J Neurosci. 2013;33:1116–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Carreno FR, Donegan JJ, Boley AM, Shah A, DeGuzman M, Frazer A, et al. Activation of a ventral hippocampus-medial prefrontal cortex pathway is both necessary and sufficient for an antidepressant response to ketamine. Mol Psychiatry. 2015;21:1298–308.

    PubMed  Google Scholar 

  67. Son H, Baek JH, Go BS, Jung DH, Sontakke SB, Chung HJ, et al. Glutamine has antidepressive effects through increments of glutamate and glutamine levels and glutamatergic activity in the medial prefrontal cortex. Neuropharmacology. 2018;143:143–52.

    CAS  PubMed  Google Scholar 

  68. Carlson D, David LK, Gallagher NM, Vu MT, Shirley M, Hultman R, et al. Dynamically timed stimulation of corticolimbic circuitry activates a stress-compensatory pathway. Biol Psychiatry. 2017;82:904–13.

    PubMed  PubMed Central  Google Scholar 

  69. Miller OH, Bruns A, Ben Ammar I, Mueggler T, Hall BJ. Synaptic regulation of a thalamocortical circuit controls depression-related behavior. Cell Rep. 2017;20:1867–80.

    CAS  PubMed  Google Scholar 

  70. Tye KM, Mirzabekov JJ, Warden MR, Ferenczi EA, Tsai H-C, Finkelstein J, et al. Dopamine neurons modulate neural encoding and expression of depression-related behaviour. Nature. 2013;493:537–41.

    CAS  PubMed  Google Scholar 

  71. Hames JL, Hagan CR, Joiner TE. Interpersonal processes in depression. Annu Rev Clin Psychol. 2013;9:355–77.

    PubMed  Google Scholar 

  72. Challis C, Berton O. Top-down control of serotonin systems by the prefrontal cortex: a path toward restored socioemotional function in depression. ACS Chem Neurosci. 2015;6:1040–54.

    CAS  PubMed  Google Scholar 

  73. Golden SA, Covington HE III, Berton O, Russo SJ. A standardized protocol for repeated social defeat stress in mice. Nat Protoc. 2011;6:1183–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Dolzani SD, Baratta MV, Moss JM, Leslie NL, Tilden SG, Sorensen AT, et al. Inhibition of a descending prefrontal circuit prevents ketamine-induced stress resilience in females. eNeuro. 2018;5:1–18.

  75. Covington HE III, Lobo MK, Maze I, Vialou V, Hyman JM, Zaman S, et al. Antidepressant effect of optogenetic stimulation of the medial prefrontal cortex. J Neurosci. 2010;30:16082–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Lee E, Hong J, Park YG, Chae S, Kim Y, Kim D. Left brain cortical activity modulates stress effects on social behavior. Sci Rep. 2015;5:13342.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Ferenczi EA, Zalocusky KA, Liston C, Grosenick L, Warden MR, Amatya D, et al. Prefrontal cortical regulation of brainwide circuit dynamics and reward-related behavior. Science. 2016;351:aac9698.

    PubMed  PubMed Central  Google Scholar 

  78. Yizhar O, Fenno LE, Prigge M, Schneider F, Davidson TJ, O’Shea DJ, et al. Neocortical excitation/inhibition balance in information processing and social dysfunction. Nature. 2011;477:171–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Warthen DM, Lambeth PS, Ottolini M, Shi Y, Barker BS, Gaykema RP, et al. Activation of pyramidal neurons in mouse medial prefrontal cortex enhances food-seeking behavior while reducing impulsivity in the absence of an effect on food intake. Front Behav Neurosci. 2016;10:63.

    PubMed  PubMed Central  Google Scholar 

  80. Felix-Ortiz AC, Burgos-Robles A, Bhagat ND, Leppla CA, Tye KM. Bidirectional modulation of anxiety-related and social behaviors by amygdala projections to the medial prefrontal cortex. Neuroscience. 2016;321:197–09.

    CAS  PubMed  Google Scholar 

  81. Liu RJ, Ota KT, Dutheil S, Duman RS, Aghajanian GK. Ketamine strengthens crf-activated amygdala inputs to basal dendrites in mpfc layer v pyramidal cells in the prelimbic but not infralimbic subregion, a key suppressor of stress responses. Neuropsychopharmacology. 2015;40:2066–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Chaudhury D, Walsh JJ, Friedman AK, Juarez B, Ku SM, Koo JW, et al. Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons. Nature. 2013;493:532–6.

    CAS  PubMed  Google Scholar 

  83. Venzala E, Garcia-Garcia AL, Elizalde N, Tordera RM. Social vs. environmental stress models of depression from a behavioural and neurochemical approach. Eur Neuropsychopharmacol. 2013;23:697–708.

    CAS  PubMed  Google Scholar 

  84. Tanaka K, Furuyashiki T, Kitaoka S, Senzai Y, Imoto Y, Segi-Nishida E, et al. Prostaglandin E2-mediated attenuation of mesocortical dopaminergic pathway is critical for susceptibility to repeated social defeat stress in mice. J Neurosci. 2012;32:4319–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Shinohara R, Taniguchi M, Ehrlich AT, Yokogawa K, Deguchi Y, Cherasse Y, et al. Dopamine D1 receptor subtype mediates acute stress-induced dendritic growth in excitatory neurons of the medial prefrontal cortex and contributes to suppression of stress susceptibility in mice. Mol Psychiatry. 2017;23:1717–30.

    PubMed  Google Scholar 

  86. Hultman R, Mague SD, Li Q, Katz BM, Michel N, Lin L, et al. Dysregulation of prefrontal cortex-mediated slow-evolving limbic dynamics drives stress-induced emotional pathology. Neuron. 2016;91:439–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Bagot RC, Parise EM, Pena CJ, Zhang HX, Maze I, Chaudhury D, et al. Ventral hippocampal afferents to the nucleus accumbens regulate susceptibility to depression. Nat Commun. 2015;6:7062.

    CAS  PubMed  Google Scholar 

  88. Challis C, Beck SG, Berton O. Optogenetic modulation of descending prefrontocortical inputs to the dorsal raphe bidirectionally bias socioaffective choices after social defeat. Front Behav Neurosci. 2014;8:43.

    PubMed  PubMed Central  Google Scholar 

  89. Challis C, Boulden J, Veerakumar A, Espallergues J, Vassoler FM, Pierce RC, et al. Raphe GABAergic neurons mediate the acquisition of avoidance after social defeat. J Neurosci. 2013;33:13978–88. 13988a

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Amat J, Baratta MV, Paul E, Bland ST, Watkins LR, Maier SF. Medial prefrontal cortex determines how stressor controllability affects behavior and dorsal raphe nucleus. Nat Neurosci. 2005;8:365–71.

    CAS  PubMed  Google Scholar 

  91. Baratta MV, Zarza CM, Gomez DM, Campeau S, Watkins LR, Maier SF. Selective activation of dorsal raphe nucleus-projecting neurons in the ventral medial prefrontal cortex by controllable stress. Eur J Neurosci. 2009;30:1111–6.

    PubMed  PubMed Central  Google Scholar 

  92. Hammack SE, Cooper MA, Lezak KR. Overlapping neurobiology of learned helplessness and conditioned defeat: implications for PTSD and mood disorders. Neuropharmacology. 2012;62:565–75.

    CAS  PubMed  Google Scholar 

  93. Morrison KE, Curry DW, Cooper MA. Social status alters defeat-induced neural activation in Syrian hamsters. Neuroscience. 2012;210:168–78.

    CAS  PubMed  Google Scholar 

  94. Kollack-Walker S, Watson SJ, Akil H. Social stress in hamsters: defeat activates specific neurocircuits within the brain. J Neurosci. 1997;17:8842–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Kessler RC, Merikangas KR, Wang PS. Prevalence, comorbidity, and service utilization for mood disorders in the United States at the beginning of the twenty-first century. Annu Rev Clin Psychol. 2007;3:137–58.

    PubMed  Google Scholar 

  96. Pellow S, Chopin P, File SE, Briley M. Validation of open:closed arm entries in an elevated plus-maze as a measure of anxiety in the rat. J Neurosci Methods. 1985;14:149–67.

    CAS  PubMed  Google Scholar 

  97. Walf AA, Frye CA. The use of the elevated plus maze as an assay of anxiety-related behavior in rodents. Nat Protoc. 2007;2:322–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Adhikari A, Lerner TN, Finkelstein J, Pak S, Jennings JH, Davidson TJ, et al. Basomedial amygdala mediates top-down control of anxiety and fear. Nature. 2015;527:179–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Adhikari A, Topiwala MA, Gordon JA. Single units in the medial prefrontal cortex with anxiety-related firing patterns are preferentially influenced by ventral hippocampal activity. Neuron. 2011;71:898–910.

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Padilla-Coreano N, Bolkan SS, Pierce GM, Blackman DR, Hardin WD, Garcia-Garcia AL, et al. Direct ventral hippocampal-prefrontal input is required for anxiety-related neural activity and behavior. Neuron. 2016;89:857–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Soumier A, Sibille E. Opposing effects of acute versus chronic blockade of frontal cortex somatostatin-positive inhibitory neurons on behavioral emotionality in mice. Neuropsychopharmacology. 2014;39:2252–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Perova Z, Delevich K, Li B. Depression of excitatory synapses onto parvalbumin interneurons in the medial prefrontal cortex in susceptibility to stress. J Neurosci. 2015;35:3201–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Nabavi S, Fox R, Proulx CD, Lin JY, Tsien RY, Malinow R. Engineering a memory with LTD and LTP. Nature. 2014;511:348–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Yun S, Reynolds RP, Petrof I, White A, Rivera PD, Segev A, et al. Stimulation of entorhinal cortex-dentate gyrus circuitry is antidepressive. Nat Med. 2018;24:658–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Creed M, Pascoli VJ, Luscher C. Addiction therapy. Refining deep brain stimulation to emulate optogenetic treatment of synaptic pathology. Science. 2015;347:659–64.

    CAS  PubMed  Google Scholar 

  106. Luscher C, Pascoli V, Creed M. Optogenetic dissection of neural circuitry: from synaptic causalities to blue prints for novel treatments of behavioral diseases. Curr Opin Neurobiol. 2015;35:95–100.

    CAS  PubMed  Google Scholar 

  107. Barthas F, Humo M, Gilsbach R, Waltisperger E, Karatas M, Leman S, et al. Cingulate overexpression of mitogen-activated protein kinase phosphatase-1 as a key factor for depression. Biol Psychiatry. 2017;82:370–9.

    CAS  PubMed  Google Scholar 

  108. Barthas F, Sellmeijer J, Hugel S, Waltisperger E, Barrot M, Yalcin I. The anterior cingulate cortex is a critical hub for pain-induced depression. Biol Psychiatry. 2015;77:236–45.

    PubMed  Google Scholar 

  109. Moghaddam B, Adams B, Verma A, Daly D. Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. J Neurosci. 1997;17:2921–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Gee S, Ellwood I, Patel T, Luongo F, Deisseroth K, Sohal VS. Synaptic activity unmasks dopamine D2 receptor modulation of a specific class of layer V pyramidal neurons in prefrontal cortex. J Neurosci. 2012;32:4959–71.

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Arnsten AF. Stress weakens prefrontal networks: molecular insults to higher cognition. Nat Neurosci. 2015;18:1376–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Anderson EM, Gomez D, Caccamise A, McPhail D, Hearing M. Chronic unpredictable stress promotes cell-specific plasticity in prefrontal cortex D1 and D2 pyramidal neurons. Neurobiol Stress. 2019;10:100152.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Schutter DJ. Antidepressant efficacy of high-frequency transcranial magnetic stimulation over the left dorsolateral prefrontal cortex in double-blind sham-controlled designs: a meta-analysis. Psycholog Med. 2009;39:65–75.

    CAS  Google Scholar 

  114. Aguirre I, Carretero B, Ibarra O, Kuhalainen J, Martinez J, Ferrer A, et al. Age predicts low-frequency transcranial magnetic stimulation efficacy in major depression. J Affect Disord. 2011;130:466–9.

    PubMed  Google Scholar 

  115. Pallanti S, Bernardi S, Di Rollo A, Antonini S, Quercioli L. Unilateral low frequency versus sequential bilateral repetitive transcranial magnetic stimulation: is simpler better for treatment of resistant depression? Neuroscience. 2010;167:323–8.

    CAS  PubMed  Google Scholar 

  116. Schutter DJ. Quantitative review of the efficacy of slow-frequency magnetic brain stimulation in major depressive disorder. Psychological Med. 2010;40:1789–95.

    CAS  Google Scholar 

  117. Duman RS, Shinohara R, Fogaca MV, Hare B. Neurobiology of rapid-acting antidepressants: convergent effects on GluA1-synaptic function. Mol Psychiatry. 2019.

  118. Kumar S, Hultman R, Hughes D, Michel N, Katz BM, Dzirasa K. Prefrontal cortex reactivity underlies trait vulnerability to chronic social defeat stress. Nat Commun. 2014;5:4537.

    CAS  PubMed  Google Scholar 

  119. Hare BD, Ghosal S, Duman RS. Rapid acting antidepressants in chronic stress models: molecular and cellular mechanisms. Chronic Stress. 2017;1:2470547017697317.

    PubMed Central  Google Scholar 

  120. Amat J, Dolzani SD, Tilden S, Christianson JP, Kubala KH, Bartholomay K, et al. Previous ketamine produces an enduring blockade of neurochemical and behavioral effects of uncontrollable stress. J Neurosci. 2016;36:153–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Brachman RA, McGowan JC, Perusini JN, Lim SC, Pham TH, Faye C, et al. Ketamine as a prophylactic against stress-induced depressive-like behavior. Biol Psychiatry. 2016;79:776–86.

    CAS  PubMed  Google Scholar 

  122. McGowan JC, LaGamma CT, Lim SC, Tsitsiklis M, Neria Y, Brachman RA, et al. Prophylactic ketamine attenuates learned fear. Neuropsychopharmacology. 2017;42:1577–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  123. McGowan JC, Hill C, Mastrodonato A, LaGamma CT, Kitayev A, Brachman RA, et al. Prophylactic ketamine alters nucleotide and neurotransmitter metabolism in brain and plasma following stress. Neuropsychopharmacology. 2018;43:1813–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Friedman AK, Walsh JJ, Juarez B, Ku SM, Chaudhury D, Wang J, et al. Enhancing depression mechanisms in midbrain dopamine neurons achieves homeostatic resilience. Science. 2014;344:313–9. https://doi.org/10.1126/science.1249240.

Download references

Acknowledgements

This work was supported by the USA NIMH (RO1 MH105910-04 and RO1 MH093897-06A1 to RSD), and the USA Brain and Behavior Research Foundation (to BDH).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Brendan D. Hare.

Ethics declarations

Conflict of interest

We declare that RSD has consulted and/or received research support from Naurex, Lilly, Forest, Johnson & Johnson, Taisho, and Sunovion. The remaining authors have no competing financial interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hare, B.D., Duman, R.S. Prefrontal cortex circuits in depression and anxiety: contribution of discrete neuronal populations and target regions. Mol Psychiatry 25, 2742–2758 (2020). https://doi.org/10.1038/s41380-020-0685-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-020-0685-9

Search

Quick links