Does mismatch negativity have utility for NMDA receptor drug development in depression?

Nicholas Murphy, Marijn Lijffijt, Nithya Ramakrishnan, Bylinda Vo-Le, Brittany Vo-Le, Sidra Iqbal, Tabish Iqbal, Brittany O'Brien, Mark A Smith, Alan C Swann, Sanjay J Mathew, Nicholas Murphy, Marijn Lijffijt, Nithya Ramakrishnan, Bylinda Vo-Le, Brittany Vo-Le, Sidra Iqbal, Tabish Iqbal, Brittany O'Brien, Mark A Smith, Alan C Swann, Sanjay J Mathew

Abstract

Rapid antidepressant effects associated with ketamine have shifted the landscape for the development of therapeutics to treat major depressive disorder (MDD) from a monoaminergic to glutamatergic model. Treatment with ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, may be effective, but has many non-glutamatergic targets, and clinical and logistical problems are potential challenges. These factors underscore the importance of manipulations of binding mechanics to produce antidepressant effects without concomitant clinical side effects. This will require identification of efficient biomarkers to monitor target engagement. The mismatch negativity (MMN) is a widely used electrophysiological signature linked to the activity of NMDA receptors (NMDAR) in humans and animals and validated in pre-clinical and clinical studies of ketamine. In this review, we explore the flexibility of the MMN and its capabilities for reliable use in drug development for NMDAR antagonists in MDD. We supplement this with findings from our own research with three distinct NMDAR antagonists. The research described illustrates that there are important distinctions between the mechanisms of NMDAR antagonism, which are further crystallized when considering the paradigm used to study the MMN. We conclude that the lack of standardized methodology currently prevents MMN from being ready for common use in drug discovery. This manuscript describes data collected from the following National Institutes of Health (NIH) and Veterans Affairs (VA) studies: AV-101, NCT03583554; lanicemine, NCT03166501; ketamine, NCT02556606.

Conflict of interest statement

VistaGen Therapeutics provided the AV-101 and placebo capsules and analyzed AV-101 metabolites (AV-101 study).

Biohaven Pharmaceuticals provided the investigational drug for the lanicemine study.

SJM has served as a consultant to Alkermes, Allergan, Axsome Therapeutics, Clexio Biosciences, Greenwich Biosciences, Intra-Cellular Therapies, Janssen, Neurocrine, Perception Neurosciences, Praxis Precision Medicines, and Sage Therapeutics. He has received research support from Biohaven Pharmaceuticals and VistaGen Therapeutics. ML has served as principal investigator for trials funded by NeuroRx and Vistagen Therapeutics and has received financial support from the MEDVAMC and the Department of Defense. MAS is an employee of VistaGen Therapeutics. The authors report no conflicts of interest.

Figures

Figure 1. Outline of the cortical response…
Figure 1. Outline of the cortical response to the generic mismatch negativity (MMN) paradigm. A) The schematic demonstrates the frequency and nature of an oddball stimulus over time. B) The solid black trace represents the grand average of the standard trials contrasted with the grand average of the deviant trials (dashed line) and the difference between them (deviant minus standard, solid red line). The MMN appears within the 150 to 220 ms window and reflects the extent of the deviation between the two conditions. C) Topographic response to each condition as recorded using a 64-channel EEG cap (10-20 montage).
Figure 2. Schematic depiction of an NMDA…
Figure 2. Schematic depiction of an NMDA receptor (NMDAR). Mechanisms of NMDAR antagonists can include blockade of subunits NR2B and NR1 (competitive antagonists), blockade of the glycine binding site (glycine antagonists), blockade of allosteric sites (noncompetitive antagonist), and blockade of the pore (uncompetitive channel blockers). The MMN appears to be affected by all types of NMDAR antagonists, indicating a generic role of NMDAR activation within its circuitry. Lanicemine and ketamine are channel blockers with different trapping block profiles. Ketamine exhibits the strongest and lanicemine the weakest trapping block. AV-101 (4-chlorokynurenine [4-Cl-KYN]) blocks NMDAR activity through competitive antagonism of the glycine-binding site.
Figure 3. Summary of the mismatch negativity…
Figure 3. Summary of the mismatch negativity (MMN) findings from the AV-101 (4-chlorokynurenine [4-Cl-KYN]) investigation. The MMN is visualized as a waveform corresponding to the difference between the standard and the deviant event-related potential (ERP) waveforms (red). The plots show data averaged across the measurement time points for the placebo group, low-dose group, and high-dose group (right). Black = standard ERP; blue = deviant ERP.
Figure 4. Summary of the mismatch negativity…
Figure 4. Summary of the mismatch negativity (MMN) findings from the lanicemine investigation. The MMN is visualized as a waveform corresponding to the difference between the standard and the deviant event-related potential (ERP) waveforms (red). The plots show data averaged across the measurement time points for the placebo group (left) and lanicemine group (right). Black = standard ERP; blue = deviant ERP.
Figure 5. Summary of the mismatch negativity…
Figure 5. Summary of the mismatch negativity (MMN) findings from the ketamine investigation. The MMN is visualized as a waveform corresponding to the difference between the standard and the deviant event-related potential (ERP) waveforms (red). The plots show data averaged across the measurement time points for the placebo group (left) and ketamine group (right). Black = standard ERP; blue = deviant ERP.

References

    1. Williams LM. Precision psychiatry: a neural circuit taxonomy for depression and anxiety. Lancet Psychiatry. 2016;3:472–80.
    1. Cole MW, Bassett DS, Power JD, Braver TS, Petersen SE. Intrinsic and task-evoked network architectures of the human brain. Neuron. 2014;83:238–51.
    1. Reiner A, Levitz J. Glutamatergic signaling in the central nervous system: ionotropic and metabotropic receptors in concert. Neuron. 2018;98:1080–98.
    1. 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.
    1. 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.
    1. Murrough JW. Ketamine as a novel antidepressant: from synapse to behavior. Clin Pharmacol Ther. 2012;91:303–9.
    1. Zanos P, Moaddel R, Morris PJ, Riggs LM, Highland JN, Georgiou P, et al. Ketamine and ketamine metabolite pharmacology: insights into therapeutic mechanisms. Pharmacol Rev. 2018;70:621–60.
    1. DeLorenzo C, DellaGioia N, Bloch M, Sanacora G, Nabulsi N, Abdallah C, et al. In vivo ketamine-induced changes in [11C] ABP688 binding to metabotropic glutamate receptor subtype 5. Biol Psychiatry. 2015;77:266–75.
    1. Stone JM, Dietrich C, Edden R, Mehta MA, De Simoni S, Reed LJ, et al. Ketamine effects on brain GABA and glutamate levels with 1H-MRS: relationship to ketamine-induced psychopathology. Mol Psychiatry. 2012;17:664–5.
    1. Abdallah CG, Jackowski A, Salas R, Gupta S, Sato JR, Mao X, et al. The nucleus accumbens and ketamine treatment in major depressive disorder. Neuropsychopharmacology. 2017;42:1739–46.
    1. Abdallah CG, De Feyter HM, Averill LA, Jiang L, Averill CL, Chowdhury GM, et al. The effects of ketamine on prefrontal glutamate neurotransmission in healthy and depressed subjects. Neuropsychopharmacology. 2018;43:2154–60.
    1. Abdallah CG, Sanacora G, Duman RS, Krystal JH. The neurobiology of depression, ketamine and rapid-acting antidepressants: is it glutamate inhibition or activation? Pharmacol Ther. 2018;190:148–58.
    1. Taylor AM, Bus T, Sprengel R, Seeburg PH, Rawlins JN, Bannerman DM. Hippocampal NMDA receptors are important for behavioural inhibition but not for encoding associative spatial memories. Philos Trans R Soc Lond B Biol Sci. 2013;369:20130149.
    1. Nikiforuk A, Popik P, Drescher KU, van Gaalen M, Relo A-L, Mezler M, et al. Effects of a positive allosteric modulator of group II metabotropic glutamate receptors, LY487379, on cognitive flexibility and impulsive-like responding in rats. J Pharmacol Exp Ther. 2010;335:665–73.
    1. Cartmell J, Schoepp DD. Regulation of neurotransmitter release by metabotropic glutamate receptors. J Neurochem. 2000;75:889–907.
    1. Rajkumar R, Farrher E, Mauler J, Sripad P, Brambilla CR, Kops ER, et al. Comparison of EEG microstates with resting state fMRI and FDG‐PET measures in the default mode network via simultaneously recorded trimodal (PET/MR/EEG) data. Hum Brain Mapp. 2018 Oct 27; doi: 10.1002/hbm.24429. doi: . Online ahead of print.
    1. Mumtaz W, Malik AS, Yasin MA, Xia L. Review on EEG and ERP predictive biomarkers for major depressive disorder. Biomed Signal Process Control. 2015;22:85–98.
    1. Buzsáki G, Draguhn A. Neuronal oscillations in cortical networks. Science. 2004;304:1926–9.
    1. O’Donnell BF, Vohs JL, Krishnan GP, Rass O, Hetrick WP, Morzorati SL. The auditory steady-state response (ASSR): a translational biomarker for schizophrenia. Suppl Clin Neurophysiol. 2013;62:101–12.
    1. Murphy N, Ramakrishnan N, Walker CP, Polizzotto NR, Cho RY. Intact auditory cortical cross-frequency coupling in early and chronic schizophrenia. Front Psychiatry. 2020;11:507.
    1. Balu DT, Coyle JT. The NMDA receptor ‘glycine modulatory site’ in schizophrenia: D-serine, glycine, and beyond. Curr Opin Pharmacol. 2015;20:109–15.
    1. Sivarao DV, Chen P, Senapati A, Yang Y, Fernandes A, Benitex Y, et al. 40 Hz auditory steady-state response is a pharmacodynamic biomarker for cortical NMDA receptors. Neuropsychopharmacology. 2016;41:2232–40.
    1. He W, Chai H, Zheng L, Yu W, Chen W, Li J, et al. Mismatch negativity in treatment-resistant depression and borderline personality disorder. Prog Neuro-Psychopharmacology Biol Psychiatry. 2010;34:366–71.
    1. Ehrlichman RS, Maxwell CR, Majumdar S, Siegel SJ. Deviance-elicited changes in event-related potentials are attenuated by ketamine in mice. J Cogn Neurosci. 2008;20:1403–14.
    1. Ahnaou A, Huysmans H, Biermans R, Manyakov NV, Drinkenburg WH. Ketamine: differential neurophysiological dynamics in functional networks in the rat brain. Transl Psychiatry. 2017;7:e1237.
    1. Rosburg T, Kreitschmann-Andermahr I. The effects of ketamine on the mismatch negativity (MMN) in humans--a meta-analysis. Clin Neurophysiol. 2016;127:1387–94.
    1. Javitt DC, Steinschneider M, Schroeder CE, Arezzo JC. Role of cortical N-methyl-D-aspartate receptors in auditory sensory memory and mismatch negativity generation: implications for schizophrenia. Proc Natl Acad Sci U S A. 1996;93:11962–7.
    1. Nagai T, Kirihara K, Tada M, Koshiyama D, Koike S, Suga M, et al. Reduced mismatch negativity is associated with increased plasma level of glutamate in first-episode psychosis. Sci Rep. 2017;7:2258.
    1. Zhou Z, Zhu H, Chen L. Effect of aripiprazole on mismatch negativity (MMN) in schizophrenia. PLoS One. 2013;8:e52186.
    1. Winkler I, Karmos G, Näätänen R. Adaptive modeling of the unattended acoustic environment reflected in the mismatch negativity event-related potential. Brain Res. 1996;742:239–52.
    1. Zhang J, Dong X, Wang L, Zhao L, Weng Z, Zhang T, et al. Gender differences in global functional connectivity during facial emotion processing: a visual MMN study. Front Behav Neurosci. 2018;12:220.
    1. Näätänen R. The perception of speech sounds by the human brain as reflected by the mismatch negativity (MMN) and its magnetic equivalent (MMNm) Psychophysiology. 2001;38:1–21.
    1. Escera C, Alho K, Winkler I, Näätänen R. Neural mechanisms of involuntary attention to acoustic novelty and change. J Cogn Neurosci. 1998;10:590–604.
    1. Schröger E. Measurement and interpretation of the mismatch negativity. Behav Res Methods Instrum Comput. 1998;30:131–45.
    1. Todd J, Heathcote A, Mullens D, Whitson LR, Provost A, Winkler I. What controls gain in gain control? Mismatch negativity (MMN), priors and system biases. Brain Topogr. 2014;27:578–89.
    1. Lieder F, Daunizeau J, Garrido MI, Friston KJ, Stephan KE. Modelling trial-by-trial changes in the mismatch negativity. PLoS Comput Biol. 2013;9:e1002911.
    1. Lieder F, Stephan KE, Daunizeau J, Garrido MI, Friston KJ. A neurocomputational model of the mismatch negativity. PLoS Comput Biol. 2013;9:e1003288.
    1. Garrido MI, Kilner JM, Stephan KE, Friston KJ. The mismatch negativity: a review of underlying mechanisms. Clin Neurophysiol. 2009;120:453–63.
    1. Stephan KE, Baldeweg T, Friston KJ. Synaptic plasticity and dysconnection in schizophrenia. Biol Psychiatry. 2006;59:929–39.
    1. Schmidt A, Diaconescu AO, Kometer M, Friston KJ, Stephan KE, Vollenweider FX. Modeling ketamine effects on synaptic plasticity during the mismatch negativity. Cereb Cortex. 2013;23:2394–406.
    1. Faber ES, Sah P. Ca2+‐activated K+ (BK) channel inactivation contributes to spike broadening during repetitive firing in the rat lateral amygdala. J Physiol. 2003;552:483–97.
    1. Wigström H, Gustafsson B. A possible correlate of the postsynaptic condition for long-lasting potentiation in the guinea pig hippocampus in vitro. Neurosci Lett. 1984;44:327–32.
    1. Harris EW, Ganong AH, Cotman CW. Long-term potentiation in the hippocampus involves activation of N-methyl-D-aspartate receptors. Brain Res. 1984;323:132–7.
    1. Harms L, Fulham WR, Todd J, Meehan C, Schall U, Hodgson DM, et al. Late deviance detection in rats is reduced, while early deviance detection is augmented by the NMDA receptor antagonist MK-801. Schizophr Res. 2018;191:43–50.
    1. Collingridge GL. Long term potentiation in the hippocampus: mechanisms of initiation and modulation by neurotransmitters. Trends Pharmacol Sci. 1985;6:407–11.
    1. Phillips JL, Norris S, Talbot J, Hatchard T, Ortiz A, Birmingham M, et al. Single and repeated ketamine infusions for reduction of suicidal ideation in treatment-resistant depression. Neuropsychopharmacology. 2020;45:606–12.
    1. Singh JB, Fedgchin M, Daly EJ, De Boer P, Cooper K, Lim P, et al. A double-blind, randomized, placebo-controlled, dose-frequency study of intravenous ketamine in patients with treatment-resistant depression. Am J Psychiatry. 2016;173:816–26.
    1. Umbricht D, Schmid L, Koller R, Vollenweider FX, Hell D, Javitt DC. Ketamine-induced deficits in auditory and visual context-dependent processing in healthy volunteers: implications for models of cognitive deficits in schizophrenia. Arch Gen Psychiatry. 2000;57:1139–47.
    1. Kreitschmann-Andermahr I, Rosburg T, Demme U, Gaser E, Nowak H, Sauer H. Effect of ketamine on the neuromagnetic mismatch field in healthy humans. Brain Res Cogn Brain Res. 2001;12:109–16.
    1. Heekeren K, Daumann J, Neukirch A, Stock C, Kawohl W, Norra C, et al. Mismatch negativity generation in the human 5HT 2A agonist and NMDA antagonist model of psychosis. Psychopharmacology (Berl) 2008;199:77–88.
    1. Gunduz-Bruce H, Reinhart RM, Roach BJ, Gueorguieva R, Oliver S, D’Souza DC, et al. Glutamatergic modulation of auditory information processing in the human brain. Biol Psychiatry. 2012;71:969–77.
    1. Featherstone RE, Shin R, Kogan JH, Liang Y, Matsumoto M, Siegel SJ. Mice with subtle reduction of NMDA NR1 receptor subunit expression have a selective decrease in mismatch negativity: implications for schizophrenia prodromal population. Neurobiol Dis. 2015;73:289–95.
    1. Witten L, Oranje B, Mørk A, Steiniger-Brach B, Glenthøj BY, Bastlund JF. Auditory sensory processing deficits in sensory gating and mismatch negativity-like responses in the social isolation rat model of schizophrenia. Behav Brain Res. 2014;266:85–93.
    1. Salisbury DF, Shenton ME, Griggs CB, Bonner-Jackson A, McCarley RW. Mismatch negativity in chronic schizophrenia and first-episode schizophrenia. Arch Gen Psychiatry. 2002;59:686–94.
    1. Light GA, Braff DL. Mismatch negativity deficits are associated with poor functioning in schizophrenia patients. Arch Gen Psychiatry. 2005;62:127–36.
    1. Umbricht D, Krljes S. Mismatch negativity in schizophrenia: a meta-analysis. Schizophr Res. 2005;76:1–23.
    1. Catts VS, Lai YL, Weickert CS, Weickert TW, Catts S V. A quantitative review of the postmortem evidence for decreased cortical N-methyl-d-aspartate receptor expression levels in schizophrenia: how can we link molecular abnormalities to mismatch negativity deficits? Biol Psychol. 2016;116:57–67.
    1. Tikhonravov D, Neuvonen T, Pertovaara A, Savioja K, Ruusuvirta T, Näätänen R, et al. Effects of an NMDA-receptor antagonist MK-801 on an MMN-like response recorded in anesthetized rats. Brain Res. 2008;1203:97–102.
    1. Sivarao DV, Chen P, Yang Y, Li YW, Pieschl R, Ahlijanian MK. NR2B antagonist CP-101,606 abolishes pitch-mediated deviance detection in awake rats. Front Psychiatry. 2014;5:96.
    1. Pochwat B, Szewczyk B, Sowa-Kucma M, Siwek A, Doboszewska U, Piekoszewski W, et al. Antidepressant-like activity of magnesium in the chronic mild stress model in rats: alterations in the NMDA receptor subunits. Int J Neuropsychopharmacol. 2014;17:393–405.
    1. Javitt DC, Lee M, Kantrowitz JT, Martinez A. Mismatch negativity as a biomarker of theta band oscillatory dysfunction in schizophrenia. Schizophr Res. 2018;191:51–60.
    1. Lee M, Sehatpour P, Hoptman MJ, Lakatos P, Dias EC, Kantrowitz JT, et al. Neural mechanisms of mismatch negativity dysfunction in schizophrenia. Mol Psychiatry. 2017;22:1585–93.
    1. Hochberger WC, Joshi YB, Thomas ML, Zhang W, Bismark AW, Treichler EB, et al. Neurophysiologic measures of target engagement predict response to auditory-based cognitive training in treatment refractory schizophrenia. Neuropsychopharmacology. 2019;44:606–12.
    1. Swerdlow NR, Bhakta S, Chou HH, Talledo JA, Balvaneda B, Light GA. Memantine effects on sensorimotor gating and mismatch negativity in patients with chronic psychosis. Neuropsychopharmacology. 2016;41:419–30.
    1. Driesen NR, McCarthy G, Bhagwagar Z, Bloch MH, Calhoun VD, D’Souza DC, et al. The impact of NMDA receptor blockade on human working memory-related prefrontal function and connectivity. Neuropsychopharmacology. 2013;38:2613–22.
    1. Murphy N, Ramakrishnan N, Vo-Le B, Vo-Le B, Smith MA, Iqbal T, et al. Electrophysiological correlates of AV-101 N-methyl-D-aspartate receptor blockade in healthy young military veterans. Neuropsychopharmacology. 2021 Forthcoming.
    1. Thomas SJ, Grossberg GT. Memantine: a review of studies into its safety and efficacy in treating Alzheimer’s disease and other dementias. Clin Interv Aging. 2009;4:367–77.
    1. Amidfar M, Réus GZ, Quevedo J, Kim YK. The role of memantine in the treatment of major depressive disorder: clinical efficacy and mechanisms of action. Eur J Pharmacol. 2018;827:103–11.
    1. Mullen T. CleanLine EEGLAB plugin. San Diego: Neuroimaging Informatics Tools and Resour Clear; 2012.
    1. Delorme A, Makeig S. EEGLAB: an open source toolbox for analysis of single-trial EEG dynamics including independent component analysis. J Neurosci Methods. 2004;134:9–21.
    1. Lijffijt M, Green CE, Balderston N, Iqbal T, Atkinson M, Vo-Le B, et al. A proof-of-mechanism study to test effects of the NMDA receptor antagonist lanicemine on behavioral sensitization in individuals with symptoms of PTSD. Front Psychiatry. 2019;10:846.
    1. Almeida OP, Hankey GJ, Yeap BB, Golledge J, Flicker L. Depression as a modifiable factor to decrease the risk of dementia. Transl Psychiatry. 2017;7:e1117.
    1. Brundin L, Bryleva EY, Rajamani KT. Role of inflammation in suicide: from mechanisms to treatment. Neuropsychopharmacology. 2017;42:271–83.
    1. Mealing GAR, Lanthorn TH, Murray CL, Small DL, Morley P. Differences in degree of trapping of low-affinity uncompetitive N-methyl-D-aspartic acid receptor antagonists with similar kinetics of block. J Pharmacol Exp Ther. 1999;288:204–10.
    1. O’Brien B, Green CE, Al-Jurdi R, Chang L, Lijffijt M, Iqbal S, et al. Bayesian adaptive randomization trial of intravenous ketamine for veterans with late-life, treatment-resistant depression. Contemp Clin Trials Commun. 2019;16:100432.
    1. Sanacora G, Smith MA, Pathak S, Su HL, Boeijinga PH, McCarthy DJ, et al. Lanicemine: a low-trapping NMDA channel blocker produces sustained antidepressant efficacy with minimal psychotomimetic adverse effects. Mol Psychiatry. 2014;19:978–85.
    1. Amenedo E, Escera C. The accuracy of sound duration representation in the human brain determines the accuracy of behavioural perception. Eur J Neurosci. 2000;12:2570–4.
    1. Alain C, Woods DL, Ogawa KH. Brain indices of automatic pattern processing. Neuroreport. 1994;6:140–4.
    1. Bissonnette JN, Francis AM, Hull KM, Leckey J, Pimer L, Berrigan LI, et al. MMN-indexed auditory change detection in major depressive disorder. Clin EEG Neurosci. 2020;51:365–72.
    1. Kim S, Baek JH, Shim SH, Kwon YJ, Lee HY, Yoo JH, et al. Mismatch negativity indices and functional outcomes in unipolar and bipolar depression. Sci Rep. 2020;10:12831.
    1. Chen J, Zhang Y, Wei D, Wu X, Fu Q, Xu F, et al. Neurophysiological handover from MMN to P3a in first-episode and recurrent major depression. J Affect Disord. 2015;174:173–9.
    1. Forsyth A, McMillan R, Campbell D, Malpas G, Maxwell E, Sleigh J, et al. Comparison of local spectral modulation, and temporal correlation, of simultaneously recorded EEG/fMRI signals during ketamine and midazolam sedation. Psychopharmacology (Berl) 2018;235:3479–93.
    1. Takei Y, Kumano S, Hattori S, Uehara T, Kawakubo Y, Kasai K, et al. Preattentive dysfunction in major depression: a magnetoencephalography study using auditory mismatch negativity. Psychophysiology. 2009;46:52–61.
    1. Qiu X, Yang X, Qiao Z, Wang L, Ning N, Shi J, et al. Impairment in processing visual information at the pre-attentive stage in patients with a major depressive disorder: a visual mismatch negativity study. Neurosci Lett. 2011;491:53–7.
    1. Restuccia D, Vollono C, Scaloni L, Buccelletti F, Camardese G. Abnormality of auditory mismatch negativity in depression and its dependence on stimulus intensity. Clin EEG Neurosci. 2016;47:105–12.
    1. Qiao Z, Yu Y, Wang L, Yang X, Qiu X, Zhang C, et al. Impaired pre-attentive change detection in major depressive disorder patients revealed by auditory mismatch negativity. Psychiatry Res. 2013;211:78–84.
    1. Pang X, Xu J, Chang Y, Tang D, Zheng Y, Liu Y, et al. Mismatch negativity of sad syllables is absent in patients with major depressive disorder. PLoS One. 2014;9:e91995.
    1. Chang Y, Xu J, Shi N, Zhang B, Zhao L. Dysfunction of processing task-irrelevant emotional faces in major depressive disorder patients revealed by expression-related visual MMN. Neurosci Lett. 2010;472:33–7.
    1. Cotter DR, Pariante CM, Everall IP. Glial cell abnormalities in major psychiatric disorders: the evidence and implications. Brain Res Bull. 2001;55:585–95.
    1. Pittenger C, Duman RS. Stress, depression, and neuroplasticity: a convergence of mechanisms. Neuropsychopharmacology. 2008;33:88–109.
    1. Ono Y, Lin YT, Liu HH, Hsieh MH. Source localization of mismatch negativity response of the auditory event-related potential using SPM8 ????? 2015;53:S440–2.
    1. Avissar M, Powell F, Ilieva I, Respino M, Gunning FM, Liston C, et al. Functional connectivity of the left DLPFC to striatum predicts treatment response of depression to TMS. Brain Stimul. 2017;10:919–25.
    1. Corlier J, Carpenter LL, Wilson AC, Tirrell E, Gobin AP, Kavanaugh B, et al. The relationship between individual alpha peak frequency and clinical outcome with repetitive Transcranial magnetic stimulation (rTMS) treatment of major depressive disorder (MDD) Brain Stimul. 2019;12:1572–8.
    1. Wang Q, Tian S, Tang H, Liu X, Yan R, Hua L, et al. Identification of major depressive disorder and prediction of treatment response using functional connectivity between the prefrontal cortices and subgenual anterior cingulate: a real-world study. J Affect Disord. 2019;252:365–72.
    1. Kähkönen S, Yamashita H, Rytsälä H, Suominen K, Ahveninen J, Isometsä E. Dysfunction in early auditory processing in major depressive disorder revealed by combined MEG and EEG. J Psychiatry Neurosci. 2007;32:316–22.
    1. Naismith SL, Mowszowski L, Ward PB, Diamond K, Paradise M, Kaur M, et al. Reduced temporal mismatch negativity in late-life depression: an event-related potential index of cognitive deficit and functional disability? J Affect Disord. 2012;138:71–8.
    1. Miyazaki T, Nakajima W, Hatano M, Shibata Y, Kuroki Y, Arisawa T, et al. Visualization of AMPA receptors in living human brain with positron emission tomography. Nat Med. 2020;26:281–8.

Source: PubMed

Подписаться