Ketamine has distinct electrophysiological and behavioral effects in depressed and healthy subjects

Allison C Nugent, Elizabeth D Ballard, Todd D Gould, Lawrence T Park, Ruin Moaddel, Nancy E Brutsche, Carlos A Zarate Jr, Allison C Nugent, Elizabeth D Ballard, Todd D Gould, Lawrence T Park, Ruin Moaddel, Nancy E Brutsche, Carlos A Zarate Jr

Abstract

Ketamine's mechanism of action was assessed using gamma power from magnetoencephalography (MEG) as a proxy measure for homeostatic balance in 35 unmedicated subjects with major depressive disorder (MDD) and 25 healthy controls enrolled in a double-blind, placebo-controlled, randomized cross-over trial of 0.5 mg/kg ketamine. MDD subjects showed significant improvements in depressive symptoms, and healthy control subjects exhibited modest but significant increases in depressive symptoms for up to 1 day after ketamine administration. Both groups showed increased resting gamma power following ketamine. In MDD subjects, gamma power was not associated with the magnitude of the antidepressant effect. However, baseline gamma power was found to moderate the relationship between post-ketamine gamma power and antidepressant response; specifically, higher post-ketamine gamma power was associated with better response in MDD subjects with lower baseline gamma, with an inverted relationship in MDD subjects with higher baseline gamma. This relationship was observed in multiple regions involved in networks hypothesized to be involved in the pathophysiology of MDD. This finding suggests biological subtypes based on the direction of homeostatic dysregulation and has important implications for inferring ketamine's mechanism of action from studies of healthy controls alone.

Conflict of interest statement

Conflict of Interest

Funding for this work was supported by the Intramural Research Program at the National Institute of Mental Health, National Institutes of Health (IRP-NIMH-NIH; NCT00088699/04-M-0222/ZIA MH002857), by a NARSAD Independent Investigator Award to Dr. Zarate, and by a Brain and Behavior Mood Disorders Research Award to Dr. Zarate. Dr. Zarate is listed as a coinventor on a patent for the use of ketamine in major depression and suicidal ideation. Dr. Zarate is listed as a co-inventor on a patent for the use of (2R,6R)-hydroxynorketamine, (S)-dehydronorketamine, and other stereoisomeric dehydro and hydroxylated metabolites of (R,S)-ketamine metabolites in the treatment of depression and neuropathic pain. Dr. Zarate is listed as co-inventor on a patent application for the use of (2R,6R)-hydroxynorketamine and (2S,6S)-hydroxynorketamine in the treatment of depression, anxiety, anhedonia, suicidal ideation, and post-traumatic stress disorders; he has assigned his patent rights to the U.S. government but will share a percentage of any royalties that may be received by the government. All other authors have no conflict of interest to disclose, financial or otherwise.

Figures

Figure 1
Figure 1
Effects of Ketamine on Mood. Graphs of the marginal means derived from the linear mixed models for the Montgomery-Åsberg Depression Rating Scale (MADRS) and Clinician Administered Dissociative States Scale (CADSS) time points significant at pA) In major depressive disorder (MDD) subjects, MADRS scores demonstrated a significant main effect of drug (F1,559=140.70, p<.001; Supplementary Table S2). CADSS scores indicated significant main effects of drug (F1,414=57.66, p<.001), time (F6,380=43.61, p<0.001), and a drug by time interaction (F6,380=45.59, <0.001). CADSS scores peaked at 40 minutes post-ketamine (F1,389=326.7, p<.001); the effect of the drug was not significant at any other time point. B) In healthy controls, MADRS scores indicated significant main effects of drug (F1,328=61.87, p<0.001) and time (F9,314=14.31, p<0.001) and a significant drug by time interaction (F9,313=9.87, p<0.001). The increase in depressive symptoms was significant at 40, 80, and 120 minutes post-infusion and at Day 1. Seventeen of 24 healthy controls receiving ketamine (71%) showed an increase of at least five points on the MADRS at any time point, compared to only one of 23 healthy controls receiving placebo (4%). By Day 2, only one healthy control subject still scored above 5 on the MADRS. CADSS scores in healthy controls demonstrated significant main effects of drug (F1,274=26.21, p<0.001), time (F6,261=27.50, p<0.001), and a drug by time interaction (F6,259=26.01, p<0.001), with significant differences between ketamine and placebo observed only at the 40-minute time point.
Figure 2
Figure 2
Images of Ketamine-Day0 vs. Placebo-Day0 contrasts derived from the mixed model are shown for A) patients with major depressive disorder (MDD). Images are thresholded at a voxel-level threshold of p<0.01, which corresponds to pFDR=0.049. The same images are shown for B) healthy controls, where in order to differentiate regions of peak change, images are thresholded at a more restrictive voxel-level threshold of p<0.001, which corresponds to pFDR=0.0014. Gamma power was robustly increased in regions of the central executive network (CEN; Figure 2a,b Z=36mm), including bilateral parietal cortex, dorsomedial prefrontal cortex, and dorsolateral prefrontal cortex (DLPFC) in both groups. The MDD group exhibited increases in the insula, which is involved in the salience network (SN; Figure 2a, Z=0mm). Healthy control subjects exhibited increases in gamma power in the posterior cingulate and thalamus (Figure 2b, Z=0mm to 12mm), regions related to the default mode network (DMN). MDD subjects exhibited increases under ketamine in the inferior temporal cortex extending into the parahippocampal cortex (Figure 2a, Z=−24mm to −12mm). C) The plots of estimated marginal means from mixed models performed on regions of interest (ROIs) defined on the Ketamine-Day0 vs. Placebo-Day0 contrast collapsed across groups. Coordinates are given in Supplementary Table S7. Because these are functionally defined ROIs, effect sizes cannot be interpreted, although the general trend of MDD subjects exhibiting increases in gamma power following ketamine infusion to a level commensurate with that of the healthy controls following placebo infusion can be observed. HC: healthy control.
Figure 3
Figure 3
A) Exploratory results in major depressive disorder (MDD) patients from a mixed model examining post-infusion gamma power in the right thalamus with baseline gamma power and change in Montgomery Åsberg Depression Rating Scale (MADRS) score as a covariate. Significant main effects were noted for both baseline gamma power (F1,12=224.8, p<0.001) and MADRS response (F1,12=32.6, p<0.001), as was a significant interaction between baseline gamma power and MADRS response (F1,12=47.46, p<0.001). The predicted values are plotted versus change in MADRS score from t=−60 to t=+40 following ketamine infusion for three groups of patients stratified by baseline gamma power to visualize the interaction. Note that subgroup stratification was performed here for visualization purposes only; baseline gamma power was a continuous variable in the statistical model. B) The same data from A (above), except with change in gamma power from baseline to ketamine sessions plotted along the y-axis; note that the statistical effects are the same.

References

    1. 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 Jan;163:28–40.
    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 Aug;63:856–864.
    1. Valentine GW, Mason GF, Gomez R, Fasula M, Watzl J, Pittman B, et al. The antidepressant effect of ketamine is not associated with changes in occipital amino acid neurotransmitter content as measured by [(1)H]-MRS. Psychiatry Res. 2011 Feb 28;191:122–127.
    1. Murrough JW, Iosifescu DV, Chang LC, Al Jurdi RK, Green CE, Perez AM, et al. Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am J Psychiatry. 2013 Oct;170:1134–1142.
    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 Aug 01;173:816–826.
    1. Zarate CA, Jr, Brutsche NE, Ibrahim L, Franco-Chaves J, Diazgranados N, Cravchik A, et al. Replication of ketamine’s antidepressant efficacy in bipolar depression: a randomized controlled add-on trial. Biol Psychiatry. 2012 Jun 01;71:939–946.
    1. DiazGranados N, Ibrahim LA, Brutsche NE, Ameli R, Henter ID, Luckenbaugh DA, et al. Rapid resolution of suicidal ideation after a single infusion of an N-methyl-D-aspartate antagonist in patients with treatment-resistant major depressive disorder. J Clin Psychiatry. 2010 Dec;71:1605–1611.
    1. Murrough JW, Soleimani L, DeWilde KE, Collins KA, Lapidus KA, Iacoviello BM, et al. Ketamine for rapid reduction of suicidal ideation: a randomized controlled trial. Psychol Med. 2015 Dec;45:3571–3580.
    1. Maeng S, Zarate CA., Jr The role of glutamate in mood disorders: results from the ketamine in major depression study and the presumed cellular mechanism underlying its antidepressant effects. Curr Psychiatry Rep. 2007 Dec;9:467–474.
    1. Maeng S, Zarate CA, Jr, Du J, Schloesser RJ, McCammon J, Chen G, et al. Cellular mechanisms underlying the antidepressant effects of ketamine: role of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors. Biol Psychiatry. 2008 Feb 15;63:349–352.
    1. 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 Aug 20;329:959–964.
    1. Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF, et al. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature. 2011 Jun 15;475:91–95.
    1. Pinault D. N-methyl d-aspartate receptor antagonists ketamine and MK-801 induce wake-related aberrant gamma oscillations in the rat neocortex. Biol Psychiatry. 2008 Apr 15;63:730–735.
    1. Anderson PM, Pinault D, O’Brien TJ, Jones NC. Chronic administration of antipsychotics attenuates ongoing and ketamine-induced increases in cortical gamma oscillations. Int J Neuropsychopharmacol. 2014 Nov;17:1895–1904.
    1. Jones NC, Anderson P, Rind G, Sullivan C, van den Buuse M, O’Brien TJ. Effects of aberrant gamma frequency oscillations on prepulse inhibition. Int J Neuropsychopharmacol. 2014 Oct;17:1671–1681.
    1. Zhang Y, Yoshida T, Katz DB, Lisman JE. NMDAR antagonist action in thalamus imposes delta oscillations on the hippocampus. J Neurophysiol. 2012 Jun;107:3181–3189.
    1. Rivolta D, Heidegger T, Scheller B, Sauer A, Schaum M, Birkner K, et al. Ketamine Dysregulates the Amplitude and Connectivity of High-Frequency Oscillations in Cortical-Subcortical Networks in Humans: Evidence From Resting-State Magnetoencephalography-Recordings. Schizophr Bull. 2015 Sep;41:1105–1114.
    1. Shaw AD, Saxena N, LEJ, Hall JE, Singh KD, Muthukumaraswamy SD. Ketamine amplifies induced gamma frequency oscillations in the human cerebral cortex. Eur Neuropsychopharmacol. 2015 Aug;25:1136–1146.
    1. Ren Z, Pribiag H, Jefferson SJ, Shorey M, Fuchs T, Stellwagen D, et al. Bidirectional Homeostatic Regulation of a Depression-Related Brain State by Gamma-Aminobutyric Acidergic Deficits and Ketamine Treatment. Biol Psychiatry. 2016 Sep 15;80:457–468.
    1. Homayoun H, Moghaddam B. NMDA receptor hypofunction produces opposite effects on prefrontal cortex interneurons and pyramidal neurons. J Neurosci. 2007 Oct 24;27:11496–11500.
    1. Carlen M, Meletis K, Siegle JH, Cardin JA, Futai K, Vierling-Claassen D, et al. A critical role for NMDA receptors in parvalbumin interneurons for gamma rhythm induction and behavior. Mol Psychiatry. 2012 May;17:537–548.
    1. Miller OH, Moran JT, Hall BJ. Two cellular hypotheses explaining the initiation of ketamine’s antidepressant actions: Direct inhibition and disinhibition. Neuropharmacology. 2016 Jan;100:17–26.
    1. Zanos P, Moaddel R, Morris PJ, Georgiou P, Fischell J, Elmer GI, et al. NMDA receptor inhibition-independent antidepressant actions of a ketamine metabolite. Nature. 2016;533:481–486.
    1. Bartos M, Vida I, Jonas P. Synaptic mechanisms of synchronized gamma oscillations in inhibitory interneuron networks. Nat Rev Neurosci. 2007 Jan;8:45–56.
    1. Fries P. Neuronal gamma-band synchronization as a fundamental process in cortical computation. Annu Rev Neurosci. 2009;32:209–224.
    1. Debener S, Herrmann CS, Kranczioch C, Gembris D, Engel AK. Top-down attentional processing enhances auditory evoked gamma band activity. Neuroreport. 2003 Apr 15;14:683–686.
    1. Mulert C, Leicht G, Hepp P, Kirsch V, Karch S, Pogarell O, et al. Single-trial coupling of the gamma-band response and the corresponding BOLD signal. Neuroimage. 2010 Feb 01;49:2238–2247.
    1. Niessing J, Ebisch B, Schmidt KE, Niessing M, Singer W, Galuske RA. Hemodynamic signals correlate tightly with synchronized gamma oscillations. Science. 2005 Aug 05;309:948–951.
    1. Shmuel A, Leopold DA. Neuronal correlates of spontaneous fluctuations in fMRI signals in monkey visual cortex: Implications for functional connectivity at rest. Hum Brain Mapp. 2008 Jul;29:751–761.
    1. Kann O, Hollnagel JO, Elzoheiry S, Schneider J. Energy and Potassium Ion Homeostasis during Gamma Oscillations. Front Mol Neurosci. 2016;9:47.
    1. Gandal MJ, Sisti J, Klook K, Ortinski PI, Leitman V, Liang Y, et al. GABAB-mediated rescue of altered excitatory-inhibitory balance, gamma synchrony and behavioral deficits following constitutive NMDAR-hypofunction. Transl Psychiatry. 2012 Jul 17;2:e142.
    1. Kehrer C, Maziashvili N, Dugladze T, Gloveli T. Altered Excitatory-Inhibitory Balance in the NMDA-Hypofunction Model of Schizophrenia. Front Mol Neurosci. 2008;1:6.
    1. Duman RS, Aghajanian GK. Synaptic dysfunction in depression: potential therapeutic targets. Science. 2012 Oct 05;338:68–72.
    1. Turrigiano GG. The self-tuning neuron: synaptic scaling of excitatory synapses. Cell. 2008 Oct 31;135:422–435.
    1. Smith KS, Rudolph U. Anxiety and depression: mouse genetics and pharmacological approaches to the role of GABA(A) receptor subtypes. Neuropharmacology. 2012 Jan;62:54–62.
    1. Thompson SM, Kallarackal AJ, Kvarta MD, Van Dyke AM, LeGates TA, Cai X. An excitatory synapse hypothesis of depression. Trends Neurosci. 2015 May;38:279–294.
    1. Drevets WC. Neuroplasticity in Mood Disorders. Dialogues Clin Neurosci. 2004;6:199–216.
    1. Drevets WC, Gadde KM, Krishnan KR. Neuroimaging Studies of Mood Disorders. In: Charney DS, editor. Neurobiology of Mental Illness. 2. Oxford University Press; Oxford, UK: 2004. pp. 461–490.
    1. Cornwell BR, Salvadore G, Furey M, Marquardt CA, Brutsche NE, Grillon C, et al. Synaptic potentiation is critical for rapid antidepressant response to ketamine in treatment-resistant major depression. Biol Psychiatry. 2012 Oct 1;72:555–561.
    1. First MB, Spitzer RL, Gibbon M, Williams AR. Structured Clinical Interview for DSM-IV TR Axis I Disorders, Research Version, Patient Edition (SCID-I/P) New York: New York State Psychiatric Institute, Biometrics Research; 2001.
    1. Sackeim HA. The definition and meaning of treatment-resistant depression. J Clin Psychiatry. 2001;62( Suppl 16):10–17.
    1. Hamilton M. A rating scale for depression. J Neurol Neurosurg Psychiatry. 1960 Feb;23:56–62.
    1. Bech P, Boyer P, Germain JM, Padmanabhan K, Haudiquet V, Pitrosky B, et al. HAM-D17 and HAM-D6 sensitivity to change in relation to desvenlafaxine dose and baseline depression severity in major depressive disorder. Pharmacopsychiatry. 2010 Nov;43:271–276.
    1. Snaith RP, Hamilton M, Morley S, Humayan A, Hargreaves D, Trigwell P. A scale for the assessment of hedonic tone the Snaith-Hamilton Pleasure Scale. Br J Psychiatry. 1995 Jul;167:99–103.
    1. Gard DE, Gard MG, Kring AM, John OP. Anticipatory and consummatory components of the experience of pleasure: A scale development study. J Res Personal. 2006 Dec;40:1086–1102.
    1. Hamilton M. The assessment of anxiety states by rating. Br J Med Psychol. 1959;32:50–55.
    1. Beck AT, Beamesderfer A. Assessment of depression: the depression inventory. Mod Probl Pharmacopsychiatry. 1974;7:151–169.
    1. Blanchard EB, Jones-Alexander J, Buckley TC, Forneris CA. Psychometric properties of the PTSD Checklist (PCL) Behav Res Ther. 1996 Aug;34:669–673.
    1. Endicott J, Nee J, Harrison W, Blumenthal R. Quality of Life Enjoyment and Satisfaction Questionnaire: a new measure. Psychopharmacol Bull. 1993;29:321–326.
    1. Guy W. Clinical Global Impressions. In: Guy W, editor. ECDEU Assessment Manual for Psychopharmacology. National Institute of Mental Health; 1976.
    1. Beck AT, Kovacs M, Weissman A. Assessment of suicidal intention: the Scale for Suicide Ideation. J Consult Clin Psychol. 1979 Apr;47:343–352.
    1. McNair DM, Lorr M, Droppleman LF. Educational and Testing Service. 1971. EITS Manual for the Profile of Mood States.
    1. Aitken RC. Measurement of feelings using visual analogue scales. Proc R Soc Med. 1969 Oct;62:989–993.
    1. Bremner JD, Krystal JH, Putnam FW, Southwick SM, Marmar C, Charney DS, et al. Measurement of dissociative states with the Clinician-Administered Dissociative States Scale (CADSS) J Trauma Stress. 1998 Jan;11:125–136.
    1. Overall JE, Gorham DR. The Brief Psychiatric Rating Scale. Psychol Rep. 1962;10:799–812.
    1. Young RC, Biggs JT, Ziegler VE, Meyer DA. A rating scale for mania: reliability, validity and sensitivity. Br J Psychiatry. 1978 Nov;133:429–435.
    1. Lopes da Silva F. EEG and MEG: relevance to neuroscience. Neuron. 2013;80:1112–1128.
    1. Gramfort A, Luessi M, Larson E, Engemann DA, Strohmeier D, Brodbeck C, et al. MEG and EEG data analysis with MNE-Python. Front Neurosci. 2013 Dec 26;7:267.
    1. Cox RW. AFNI: software for analysis and visualization of functional magnetic resonance neuroimages. Comput Biomed Res. 1996 Jun;29:162–173.
    1. Robinson SE, Vrba J. Biomag. Tohoku University Press; Sendai: 1999. Functional neuroimaging by synthetic aperture magnetometry (SAM) pp. 302–305.
    1. Chen G, Saad ZS, Britton JC, Pine DS, Cox RW. Linear mixed–effects modeling approach to FMRI group analysis. Neuroimage. 2013 Jun;73:176–190.
    1. Perry EB, Jr, Cramer JA, Cho HS, Petrakis IL, Karper LP, Genovese A, et al. Psychiatric safety of ketamine in psychopharmacology research. Psychopharmacology (Berl) 2007 Jun;192:253–260.
    1. Taylor MJ, Tiangga ER, Mhuircheartaigh RN, Cowen PJ. Lack of effect of ketamine on cortical glutamate and glutamine in healthy volunteers: a proton magnetic resonance spectroscopy study. J Psychopharmacol. 2012 May;26:733–737.
    1. Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, et al. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994 Mar;51:199–214.
    1. Stone JM, Erlandsson K, Arstad E, Squassante L, Teneggi V, Bressan RA, et al. Relationship between ketamine-induced psychotic symptoms and NMDA receptor occupancy: a [(123)I]CNS-1261 SPET study. Psychopharmacology (Berl) 2008 Apr;197:401–408.
    1. Mason OJ, Morgan CJ, Stefanovic A, Curran HV. The psychotomimetic states inventory (PSI): measuring psychotic-type experiences from ketamine and cannabis. Schizophr Res. 2008 Aug;103:138–142.
    1. Pollak TA, De Simoni S, Barimani B, Zelaya FO, Stone JM, Mehta MA. Phenomenologically distinct psychotomimetic effects of ketamine are associated with cerebral blood flow changes in functionally relevant cerebral foci: a continuous arterial spin labelling study. Psychopharmacology (Berl) 2015 Dec;232:4515–4524.
    1. Muthukumaraswamy SD, Shaw AD, Jackson LE, Hall J, Moran R, Saxena N. Evidence that Subanesthetic Doses of Ketamine Cause Sustained Disruptions of NMDA and AMPA-Mediated Frontoparietal Connectivity in Humans. J Neurosci. 2015 Aug 19;35:11694–11706.
    1. Scheidegger M, Walter M, Lehmann M, Metzger C, Grimm S, Boeker H, et al. Ketamine decreases resting state functional network connectivity in healthy subjects: implications for antidepressant drug action. PLoS One. 2012;7:e44799.
    1. Li M, Demenescu LR, Colic L, Metzger CD, Heinze HJ, Steiner J, et al. Temporal Dynamics of Antidepressant Ketamine Effects on Glutamine Cycling Follow Regional Fingerprints of AMPA and NMDA Receptor Densities. Neuropsychopharmacology. 2017 May;42:1201–1209.
    1. Haegens S, Cousijn H, Wallis G, Harrison PJ, Nobre AC. Inter- and intra-individual variability in alpha peak frequency. Neuroimage. 2014 May 15;92:46–55.
    1. 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 Jan;23:28–38.
    1. Duncan NW, Wiebking C, Tiret B, Marjanska M, Hayes DJ, Lyttleton O, et al. Glutamate concentration in the medial prefrontal cortex predicts resting-state cortical-subcortical functional connectivity in humans. PLoS One. 2013;8:e60312.
    1. Kapogiannis D, Reiter DA, Willette AA, Mattson MP. Posteromedial cortex glutamate and GABA predict intrinsic functional connectivity of the default mode network. Neuroimage. 2013 Jan 1;64:112–119.
    1. Nugent AC, Martinez A, D’Alfonso A, Zarate CA, Theodore WH. The relationship between glucose metabolism, resting-state fMRI BOLD signal, and GABA-binding potential: a preliminary study in healthy subjects and those with temporal lobe epilepsy. J Cereb Blood Flow Metab. 2015 Jan 7;
    1. Tomasi D, Wang GJ, Volkow ND. Energetic cost of brain functional connectivity. Proc Natl Acad Sci U S A. 2013 Aug 13;110:13642–13647.
    1. Niciu MJ, Ionescu DF, Richards EM, Zarate CA., Jr Glutamate and its receptors in the pathophysiology and treatment of major depressive disorder. J Neural Transm (Vienna) 2014 Aug;121:907–924.
    1. Menon V. Large-scale brain networks and psychopathology: a unifying triple network model. Trends Cogn Sci. 2011 Oct;15:483–506.
    1. Zheng H, Xu L, Xie F, Guo X, Zhang J, Yao L, et al. The Altered Triple Networks Interaction in Depression under Resting State Based on Graph Theory. Biomed Res Int. 2015;2015:386326.
    1. Manoliu A, Meng C, Brandl F, Doll A, Tahmasian M, Scherr M, et al. Insular dysfunction within the salience network is associated with severity of symptoms and aberrant inter-network connectivity in major depressive disorder. Front Hum Neurosci. 2013;7:930.
    1. Pinault D, Deschenes M. Voltage-dependent 40-Hz oscillations in rat reticular thalamic neurons in vivo. Neuroscience. 1992 Nov;51:245–258.
    1. Hoflich A, Hahn A, Kublbock M, Kranz GS, Vanicek T, Windischberger C, et al. Ketamine-induced modulation of the thalamo-cortical network in healthy volunteers as a model for schizophrenia. Int J Neuropsychopharmacol. 2015 Apr;19:18. pii:pyv040.
    1. Driesen NR, McCarthy G, Bhagwagar Z, Bloch M, Calhoun V, D’Souza DC, et al. Relationship of resting brain hyperconnectivity and schizophrenia-like symptoms produced by the NMDA receptor antagonist ketamine in humans. Mol Psychiatry. 2013 Jan 22;18:1199–1204.
    1. Ribary U, Ioannides AA, Singh KD, Hasson R, Bolton JP, Lado F, et al. Magnetic field tomography of coherent thalamocortical 40-Hz oscillations in humans. Proc Natl Acad Sci U S A. 1991 Dec 15;88:11037–11041.
    1. Luo Q, Holroyd T, Jones M, Hendler T, Blair J. Neural dynamics for facial threat processing as revealed by gamma band synchronization using MEG. Neuroimage. 2007 Jan 15;34:839–847.
    1. Nugent AC, Davis RM, Zarate CA, Jr, Drevets WC. Reduced thalamic volumes in major depressive disorder. Psychiatry Res. 2013 Sep 30;213:179–185.
    1. Drevets WC, Videen TO, Price JL, Preskorn SH, Carmichael ST, Raichle ME. A functional anatomical study of unipolar depression. J Neurosci. 1992 Sep;12:3628–3641.
    1. Greicius MD, Flores BH, Menon V, Glover GH, Solvason HB, Kenna H, et al. Resting-state functional connectivity in major depression: abnormally increased contributions from subgenual cingulate cortex and thalamus. Biol Psychiatry. 2007 Sep 1;62:429–437.

Source: PubMed

3
Prenumerera