Skip to main content
Top
Published in: Brain Structure and Function 3/2017

01-04-2017 | Original Article

Ketamine-dependent neuronal activation in healthy volunteers

Authors: Anna Höflich, Andreas Hahn, Martin Küblböck, Georg S. Kranz, Thomas Vanicek, Sebastian Ganger, Marie Spies, Christian Windischberger, Siegfried Kasper, Dietmar Winkler, Rupert Lanzenberger

Published in: Brain Structure and Function | Issue 3/2017

Login to get access

Abstract

Over the last years, a number of studies have been conducted to clarify the neurobiological correlates of ketamine application. However, comprehensive information regarding the influence of ketamine on cortical activity is still lacking. Using resting-state functional MRI and integrating pharmacokinetic information, a double-blind, randomized, placebo-controlled, crossover study was performed to determine the effects of ketamine on neuronal activation. During a 55 min resting-state fMRI scan, esketamine (Ketanest S®) was administered intravenously to 35 healthy volunteers. Neural activation as indicated by the BOLD signal using the pharmacokinetic curve of ketamine plasma levels as a regressor was computed. Compared with placebo, ketamine-dependent increases of neural activation were observed in the midcingulate cortex, the dorsal part of the anterior cingulate cortex, the insula bilaterally, and the thalamus (t values ranging between 5.95–9.78, p < 0.05; FWE-corrected). A significant decrease of neural activation in the ketamine condition compared to placebo was found in a cluster within the subgenual/subcallosal part of the anterior cingulate cortex, the orbitofrontal cortex and the gyrus rectus (t = 7.81, p < 0.05, FWE-corrected). Using an approach combining pharmacological and fMRI data, important information about the neurobiological correlates of the clinical antidepressant effects of ketamine could be revealed.
Literature
go back to reference aan het Rot M, Collins KA, Murrough JW, Perez AM, Reich DL, Charney DS, Mathew SJ (2010) Safety and efficacy of repeated-dose intravenous ketamine for treatment-resistant depression. Biol Psychiatry 67(2):139–145CrossRefPubMed aan het Rot M, Collins KA, Murrough JW, Perez AM, Reich DL, Charney DS, Mathew SJ (2010) Safety and efficacy of repeated-dose intravenous ketamine for treatment-resistant depression. Biol Psychiatry 67(2):139–145CrossRefPubMed
go back to reference Abdallah CG, Averill LA, Krystal JH (2015) Ketamine as a promising prototype for a new generation of rapid-acting antidepressants. Ann N Y Acad Sci 1344:66–77CrossRefPubMedPubMedCentral Abdallah CG, Averill LA, Krystal JH (2015) Ketamine as a promising prototype for a new generation of rapid-acting antidepressants. Ann N Y Acad Sci 1344:66–77CrossRefPubMedPubMedCentral
go back to reference Anticevic A, Corlett PR, Cole MW, Savic A, Gancsos M, Tang Y, Repovs G, Murray JD, Driesen NR, Morgan PT, Xu K, Wang F, Krystal JH (2015) N-methyl-d-aspartate receptor antagonist effects on prefrontal cortical connectivity better model early than chronic schizophrenia. Biol Psychiatry 77(6):569–580CrossRefPubMed Anticevic A, Corlett PR, Cole MW, Savic A, Gancsos M, Tang Y, Repovs G, Murray JD, Driesen NR, Morgan PT, Xu K, Wang F, Krystal JH (2015) N-methyl-d-aspartate receptor antagonist effects on prefrontal cortical connectivity better model early than chronic schizophrenia. Biol Psychiatry 77(6):569–580CrossRefPubMed
go back to reference Aroni F, Iacovidou N, Dontas I, Pourzitaki C, Xanthos T (2009) Pharmacological aspects and potential new clinical applications of ketamine: reevaluation of an old drug. J Clin Pharmacol 49(8):957–964CrossRefPubMed Aroni F, Iacovidou N, Dontas I, Pourzitaki C, Xanthos T (2009) Pharmacological aspects and potential new clinical applications of ketamine: reevaluation of an old drug. J Clin Pharmacol 49(8):957–964CrossRefPubMed
go back to reference Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, Krystal JH (2000) Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 47(4):351–354CrossRefPubMed Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, Krystal JH (2000) Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 47(4):351–354CrossRefPubMed
go back to reference Borsook D, Becerra L, Hargreaves R (2006) A role for fMRI in optimizing CNS drug development. Nat Rev Drug Discov 5(5):411–424CrossRefPubMed Borsook D, Becerra L, Hargreaves R (2006) A role for fMRI in optimizing CNS drug development. Nat Rev Drug Discov 5(5):411–424CrossRefPubMed
go back to reference Bustillo JR, Rowland LM, Mullins P, Jung R, Chen H, Qualls C, Hammond R, Brooks WM, Lauriello J (2010) 1H-MRS at 4 tesla in minimally treated early schizophrenia. Mol Psychiatry 15(6):629–636CrossRefPubMed Bustillo JR, Rowland LM, Mullins P, Jung R, Chen H, Qualls C, Hammond R, Brooks WM, Lauriello J (2010) 1H-MRS at 4 tesla in minimally treated early schizophrenia. Mol Psychiatry 15(6):629–636CrossRefPubMed
go back to reference Dandash O, Harrison BJ, Adapa R, Gaillard R, Giorlando F, Wood SJ, Fletcher PC, Fornito A (2015) Selective augmentation of striatal functional connectivity following NMDA receptor antagonism: implications for psychosis. Neuropsychopharmacology 40(3):622–631CrossRefPubMed Dandash O, Harrison BJ, Adapa R, Gaillard R, Giorlando F, Wood SJ, Fletcher PC, Fornito A (2015) Selective augmentation of striatal functional connectivity following NMDA receptor antagonism: implications for psychosis. Neuropsychopharmacology 40(3):622–631CrossRefPubMed
go back to reference De Simoni S, Schwarz AJ, O’Daly OG, Marquand AF, Brittain C, Gonzales C, Stephenson S, Williams SC, Mehta MA (2013) Test–retest reliability of the BOLD pharmacological MRI response to ketamine in healthy volunteers. Neuroimage 64:75–90CrossRefPubMed De Simoni S, Schwarz AJ, O’Daly OG, Marquand AF, Brittain C, Gonzales C, Stephenson S, Williams SC, Mehta MA (2013) Test–retest reliability of the BOLD pharmacological MRI response to ketamine in healthy volunteers. Neuroimage 64:75–90CrossRefPubMed
go back to reference Deakin JF, Lees J, McKie S, Hallak JE, Williams SR, Dursun SM (2008) Glutamate and the neural basis of the subjective effects of ketamine: a pharmaco-magnetic resonance imaging study. Arch Gen Psychiatry 65(2):154–164CrossRefPubMed Deakin JF, Lees J, McKie S, Hallak JE, Williams SR, Dursun SM (2008) Glutamate and the neural basis of the subjective effects of ketamine: a pharmaco-magnetic resonance imaging study. Arch Gen Psychiatry 65(2):154–164CrossRefPubMed
go back to reference Doyle OM, De Simoni S, Schwarz AJ, Brittain C, O’Daly OG, Williams SC, Mehta MA (2013) Quantifying the attenuation of the ketamine pharmacological magnetic resonance imaging response in humans: a validation using antipsychotic and glutamatergic agents. J Pharmacol Exp Ther 345(1):151–160CrossRefPubMed Doyle OM, De Simoni S, Schwarz AJ, Brittain C, O’Daly OG, Williams SC, Mehta MA (2013) Quantifying the attenuation of the ketamine pharmacological magnetic resonance imaging response in humans: a validation using antipsychotic and glutamatergic agents. J Pharmacol Exp Ther 345(1):151–160CrossRefPubMed
go back to reference Drevets WC, Price JL, Simpson JR Jr, Todd RD, Reich T, Vannier M, Raichle ME (1997) Subgenual prefrontal cortex abnormalities in mood disorders. Nature 386(6627):824–827CrossRefPubMed Drevets WC, Price JL, Simpson JR Jr, Todd RD, Reich T, Vannier M, Raichle ME (1997) Subgenual prefrontal cortex abnormalities in mood disorders. Nature 386(6627):824–827CrossRefPubMed
go back to reference Driesen NR, McCarthy G, Bhagwagar Z, Bloch M, Calhoun V, D’Souza DC, Gueorguieva R, He G, Ramachandran R, Suckow RF, Anticevic A, Morgan PT, Krystal JH (2013) Relationship of resting brain hyperconnectivity and schizophrenia-like symptoms produced by the NMDA receptor antagonist ketamine in humans. Mol Psychiatry 18:1199–1204CrossRefPubMedPubMedCentral Driesen NR, McCarthy G, Bhagwagar Z, Bloch M, Calhoun V, D’Souza DC, Gueorguieva R, He G, Ramachandran R, Suckow RF, Anticevic A, Morgan PT, Krystal JH (2013) Relationship of resting brain hyperconnectivity and schizophrenia-like symptoms produced by the NMDA receptor antagonist ketamine in humans. Mol Psychiatry 18:1199–1204CrossRefPubMedPubMedCentral
go back to reference du Jardin KG, Liebenberg N, Muller HK, Elfving B, Sanchez C, Wegener G (2016) Differential interaction with the serotonin system by S-ketamine, vortioxetine, and fluoxetine in a genetic rat model of depression. Psychopharmacology 233(14):2813–2825CrossRefPubMed du Jardin KG, Liebenberg N, Muller HK, Elfving B, Sanchez C, Wegener G (2016) Differential interaction with the serotonin system by S-ketamine, vortioxetine, and fluoxetine in a genetic rat model of depression. Psychopharmacology 233(14):2813–2825CrossRefPubMed
go back to reference Duman RS, Li N, Liu RJ, Duric V, Aghajanian G (2012) Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology 62(1):35–41CrossRefPubMed Duman RS, Li N, Liu RJ, Duric V, Aghajanian G (2012) Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology 62(1):35–41CrossRefPubMed
go back to reference Franceschini MA, Radhakrishnan H, Thakur K, Wu W, Ruvinskaya S, Carp S, Boas DA (2010) The effect of different anesthetics on neurovascular coupling. Neuroimage 51(4):1367–1377CrossRefPubMedPubMedCentral Franceschini MA, Radhakrishnan H, Thakur K, Wu W, Ruvinskaya S, Carp S, Boas DA (2010) The effect of different anesthetics on neurovascular coupling. Neuroimage 51(4):1367–1377CrossRefPubMedPubMedCentral
go back to reference Francois J, Grimm O, Schwarz AJ, Schweiger J, Haller L, Risterucci C, Bohringer A, Zang Z, Tost H, Gilmour G, Meyer-Lindenberg A (2016) Ketamine suppresses the ventral striatal response to reward anticipation: a cross-species translational neuroimaging study. Neuropsychopharmacology 41:1386–1394CrossRefPubMed Francois J, Grimm O, Schwarz AJ, Schweiger J, Haller L, Risterucci C, Bohringer A, Zang Z, Tost H, Gilmour G, Meyer-Lindenberg A (2016) Ketamine suppresses the ventral striatal response to reward anticipation: a cross-species translational neuroimaging study. Neuropsychopharmacology 41:1386–1394CrossRefPubMed
go back to reference Geisslinger G, Hering W, Thomann P, Knoll R, Kamp HD, Brune K (1993) Pharmacokinetics and pharmacodynamics of ketamine enantiomers in surgical patients using a stereoselective analytical method. Br J Anaesth 70(6):666–671CrossRefPubMed Geisslinger G, Hering W, Thomann P, Knoll R, Kamp HD, Brune K (1993) Pharmacokinetics and pharmacodynamics of ketamine enantiomers in surgical patients using a stereoselective analytical method. Br J Anaesth 70(6):666–671CrossRefPubMed
go back to reference Gluck MR, Thomas RG, Davis KL, Haroutunian V (2002) Implications for altered glutamate and GABA metabolism in the dorsolateral prefrontal cortex of aged schizophrenic patients. Am J Psychiatry 159(7):1165–1173CrossRefPubMed Gluck MR, Thomas RG, Davis KL, Haroutunian V (2002) Implications for altered glutamate and GABA metabolism in the dorsolateral prefrontal cortex of aged schizophrenic patients. Am J Psychiatry 159(7):1165–1173CrossRefPubMed
go back to reference Habas C (2010) Functional connectivity of the human rostral and caudal cingulate motor areas in the brain resting state at 3T. Neuroradiology 52(1):47–59CrossRefPubMed Habas C (2010) Functional connectivity of the human rostral and caudal cingulate motor areas in the brain resting state at 3T. Neuroradiology 52(1):47–59CrossRefPubMed
go back to reference Hahn A, Stein P, Windischberger C, Weissenbacher A, Spindelegger C, Moser E, Kasper S, Lanzenberger R (2011) Reduced resting-state functional connectivity between amygdala and orbitofrontal cortex in social anxiety disorder. Neuroimage 56(3):881–889CrossRefPubMed Hahn A, Stein P, Windischberger C, Weissenbacher A, Spindelegger C, Moser E, Kasper S, Lanzenberger R (2011) Reduced resting-state functional connectivity between amygdala and orbitofrontal cortex in social anxiety disorder. Neuroimage 56(3):881–889CrossRefPubMed
go back to reference Hamani C, Mayberg H, Stone S, Laxton A, Haber S, Lozano AM (2011) The subcallosal cingulate gyrus in the context of major depression. Biol Psychiatry 69(4):301–308CrossRefPubMed Hamani C, Mayberg H, Stone S, Laxton A, Haber S, Lozano AM (2011) The subcallosal cingulate gyrus in the context of major depression. Biol Psychiatry 69(4):301–308CrossRefPubMed
go back to reference Hashimoto K, Kakiuchi T, Ohba H, Nishiyama S, Tsukada H (2016) Reduction of dopamine D2/3 receptor binding in the striatum after a single administration of esketamine, but not R-ketamine: a PET study in conscious monkeys. Eur Arch Psychiatry Clin Neurosci. doi:10.1007/s00406-016-0692-7 PubMedCentral Hashimoto K, Kakiuchi T, Ohba H, Nishiyama S, Tsukada H (2016) Reduction of dopamine D2/3 receptor binding in the striatum after a single administration of esketamine, but not R-ketamine: a PET study in conscious monkeys. Eur Arch Psychiatry Clin Neurosci. doi:10.​1007/​s00406-016-0692-7 PubMedCentral
go back to reference Hayton SM, Kriss A, Muller DP (1999) Comparison of the effects of four anaesthetic agents on somatosensory evoked potentials in the rat. Lab Anim 33(3):243–251CrossRefPubMed Hayton SM, Kriss A, Muller DP (1999) Comparison of the effects of four anaesthetic agents on somatosensory evoked potentials in the rat. Lab Anim 33(3):243–251CrossRefPubMed
go back to reference Himmelseher S, Durieux ME (2005) Ketamine for perioperative pain management. Anesthesiology 102(1):211–220CrossRefPubMed Himmelseher S, Durieux ME (2005) Ketamine for perioperative pain management. Anesthesiology 102(1):211–220CrossRefPubMed
go back to reference Hoflich A, Hahn A, Kublbock M, Kranz GS, Vanicek T, Windischberger C, Saria A, Kasper S, Winkler D, Lanzenberger R (2015) Ketamine-induced modulation of the thalamo-cortical network in healthy volunteers as a model for schizophrenia. Int J Neuropsychopharmacol 18(9). doi:10.1093/ijnp/pyv040 Hoflich A, Hahn A, Kublbock M, Kranz GS, Vanicek T, Windischberger C, Saria A, Kasper S, Winkler D, Lanzenberger R (2015) Ketamine-induced modulation of the thalamo-cortical network in healthy volunteers as a model for schizophrenia. Int J Neuropsychopharmacol 18(9). doi:10.​1093/​ijnp/​pyv040
go back to reference Jenkins BG (2012) Pharmacologic magnetic resonance imaging (phMRI): imaging drug action in the brain. Neuroimage 62(2):1072–1085CrossRefPubMed Jenkins BG (2012) Pharmacologic magnetic resonance imaging (phMRI): imaging drug action in the brain. Neuroimage 62(2):1072–1085CrossRefPubMed
go back to reference Kegeles L, Mao X, Stanford AD et al (2012) Elevated prefrontal cortex γ-aminobutyric acid and glutamate-glutamine levels in schizophrenia measured in vivo with proton magnetic resonance spectroscopy. Arch Gen Psychiatry 69(5):449–459CrossRefPubMed Kegeles L, Mao X, Stanford AD et al (2012) Elevated prefrontal cortex γ-aminobutyric acid and glutamate-glutamine levels in schizophrenia measured in vivo with proton magnetic resonance spectroscopy. Arch Gen Psychiatry 69(5):449–459CrossRefPubMed
go back to reference Kornhuber J, Mack-Burkhardt F, Riederer P, Hebenstreit GF, Reynolds GP, Andrews HB, Beckmann H (1989) [3H]MK-801 binding sites in postmortem brain regions of schizophrenic patients. J Neural Transm 77(2–3):231–236CrossRefPubMed Kornhuber J, Mack-Burkhardt F, Riederer P, Hebenstreit GF, Reynolds GP, Andrews HB, Beckmann H (1989) [3H]MK-801 binding sites in postmortem brain regions of schizophrenic patients. J Neural Transm 77(2–3):231–236CrossRefPubMed
go back to reference Krystal JH, Sanacora G, Duman RS (2013) Rapid-acting glutamatergic antidepressants: the path to ketamine and beyond. Biol Psychiatry 73(12):1133–1141CrossRefPubMedPubMedCentral Krystal JH, Sanacora G, Duman RS (2013) Rapid-acting glutamatergic antidepressants: the path to ketamine and beyond. Biol Psychiatry 73(12):1133–1141CrossRefPubMedPubMedCentral
go back to reference Lehmann M, Seifritz E, Henning A, Walter M, Böker H, Scheidegger M, Grimm S (2016) Differential effects of rumination and distraction on ketamine induced modulation of resting state functional connectivity and reactivity of regions within the default-mode network. Soc Cogn Affect Neurosci 11(8):1227–1235CrossRefPubMed Lehmann M, Seifritz E, Henning A, Walter M, Böker H, Scheidegger M, Grimm S (2016) Differential effects of rumination and distraction on ketamine induced modulation of resting state functional connectivity and reactivity of regions within the default-mode network. Soc Cogn Affect Neurosci 11(8):1227–1235CrossRefPubMed
go back to reference Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, Li XY, Aghajanian G, Duman RS (2010) mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science 329(5994):959–964CrossRefPubMedPubMedCentral Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, Li XY, Aghajanian G, Duman RS (2010) mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science 329(5994):959–964CrossRefPubMedPubMedCentral
go back to reference Li N, Liu RJ, Dwyer JM, Banasr M, Lee B, Son H, Li XY, Aghajanian G, Duman RS (2011) Glutamate N-methyl-d-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits caused by chronic stress exposure. Biol Psychiatry 69(8):754–761CrossRefPubMedPubMedCentral Li N, Liu RJ, Dwyer JM, Banasr M, Lee B, Son H, Li XY, Aghajanian G, Duman RS (2011) Glutamate N-methyl-d-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits caused by chronic stress exposure. Biol Psychiatry 69(8):754–761CrossRefPubMedPubMedCentral
go back to reference Lopez-Gil X, Babot Z, Amargos-Bosch M, Sunol C, Artigas F, Adell A (2007) Clozapine and haloperidol differently suppress the MK-801-increased glutamatergic and serotonergic transmission in the medial prefrontal cortex of the rat. Neuropsychopharmacology 32(10):2087–2097CrossRefPubMed Lopez-Gil X, Babot Z, Amargos-Bosch M, Sunol C, Artigas F, Adell A (2007) Clozapine and haloperidol differently suppress the MK-801-increased glutamatergic and serotonergic transmission in the medial prefrontal cortex of the rat. Neuropsychopharmacology 32(10):2087–2097CrossRefPubMed
go back to reference Lorrain DS, Baccei CS, Bristow LJ, Anderson JJ, Varney MA (2003) Effects of ketamine and N-methyl-d-aspartate on glutamate and dopamine release in the rat prefrontal cortex: modulation by a group II selective metabotropic glutamate receptor agonist LY379268. Neuroscience 117(3):697–706CrossRefPubMed Lorrain DS, Baccei CS, Bristow LJ, Anderson JJ, Varney MA (2003) Effects of ketamine and N-methyl-d-aspartate on glutamate and dopamine release in the rat prefrontal cortex: modulation by a group II selective metabotropic glutamate receptor agonist LY379268. Neuroscience 117(3):697–706CrossRefPubMed
go back to reference Margulies DS, Kelly AM, Uddin LQ, Biswal BB, Castellanos FX, Milham MP (2007) Mapping the functional connectivity of anterior cingulate cortex. Neuroimage 37(2):579–588CrossRefPubMed Margulies DS, Kelly AM, Uddin LQ, Biswal BB, Castellanos FX, Milham MP (2007) Mapping the functional connectivity of anterior cingulate cortex. Neuroimage 37(2):579–588CrossRefPubMed
go back to reference Mayberg HS, Lozano AM, Voon V, McNeely HE, Seminowicz D, Hamani C, Schwalb JM, Kennedy SH (2005) Deep brain stimulation for treatment-resistant depression. Neuron 45(5):651–660CrossRefPubMed Mayberg HS, Lozano AM, Voon V, McNeely HE, Seminowicz D, Hamani C, Schwalb JM, Kennedy SH (2005) Deep brain stimulation for treatment-resistant depression. Neuron 45(5):651–660CrossRefPubMed
go back to reference McCabe C, Mishor Z, Cowen PJ, Harmer CJ (2010) Diminished neural processing of aversive and rewarding stimuli during selective serotonin reuptake inhibitor treatment. Biol Psychiatry 67(5):439–445CrossRefPubMedPubMedCentral McCabe C, Mishor Z, Cowen PJ, Harmer CJ (2010) Diminished neural processing of aversive and rewarding stimuli during selective serotonin reuptake inhibitor treatment. Biol Psychiatry 67(5):439–445CrossRefPubMedPubMedCentral
go back to reference Murrough JW, Iosifescu DV, Chang LC, Al Jurdi RK, Green CE, Perez AM, Iqbal S, Pillemer S, Foulkes A, Shah A, Charney DS, Mathew SJ (2013a) Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am J Psychiatry 170(10):1134–1142CrossRefPubMedPubMedCentral Murrough JW, Iosifescu DV, Chang LC, Al Jurdi RK, Green CE, Perez AM, Iqbal S, Pillemer S, Foulkes A, Shah A, Charney DS, Mathew SJ (2013a) Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am J Psychiatry 170(10):1134–1142CrossRefPubMedPubMedCentral
go back to reference Murrough JW, Perez AM, Pillemer S, Stern J, Parides MK, aan het Rot M, Collins KA, Mathew SJ, Charney DS, Iosifescu DV (2013b) Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression. Biol Psychiatry 74(4):250–256CrossRefPubMed Murrough JW, Perez AM, Pillemer S, Stern J, Parides MK, aan het Rot M, Collins KA, Mathew SJ, Charney DS, Iosifescu DV (2013b) Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression. Biol Psychiatry 74(4):250–256CrossRefPubMed
go back to reference Musso F, Brinkmeyer J, Ecker D, London MK, Thieme G, Warbrick T, Wittsack HJ, Saleh A, Greb W, de Boer P, Winterer G (2011) Ketamine effects on brain function–simultaneous fMRI/EEG during a visual oddball task. Neuroimage 58(2):508–525CrossRefPubMed Musso F, Brinkmeyer J, Ecker D, London MK, Thieme G, Warbrick T, Wittsack HJ, Saleh A, Greb W, de Boer P, Winterer G (2011) Ketamine effects on brain function–simultaneous fMRI/EEG during a visual oddball task. Neuroimage 58(2):508–525CrossRefPubMed
go back to reference Nobler MS, Oquendo MA, Kegeles LS, Malone KM, Campbell CC, Sackeim HA, Mann JJ (2001) Decreased regional brain metabolism after ECT. Am J Psychiatry 158(2):305–308CrossRefPubMed Nobler MS, Oquendo MA, Kegeles LS, Malone KM, Campbell CC, Sackeim HA, Mann JJ (2001) Decreased regional brain metabolism after ECT. Am J Psychiatry 158(2):305–308CrossRefPubMed
go back to reference Ossewaarde L, Verkes RJ, Hermans EJ, Kooijman SC, Urner M, Tendolkar I, van Wingen GA, Fernandez G (2011) Two-week administration of the combined serotonin-noradrenaline reuptake inhibitor duloxetine augments functioning of mesolimbic incentive processing circuits. Biol Psychiatry 70(6):568–574CrossRefPubMed Ossewaarde L, Verkes RJ, Hermans EJ, Kooijman SC, Urner M, Tendolkar I, van Wingen GA, Fernandez G (2011) Two-week administration of the combined serotonin-noradrenaline reuptake inhibitor duloxetine augments functioning of mesolimbic incentive processing circuits. Biol Psychiatry 70(6):568–574CrossRefPubMed
go back to reference Palaniyappan L, Liddle PF (2012) Does the salience network play a cardinal role in psychosis? An emerging hypothesis of insular dysfunction. J Psychiatry Neurosci 37(1):17–27CrossRefPubMedPubMedCentral Palaniyappan L, Liddle PF (2012) Does the salience network play a cardinal role in psychosis? An emerging hypothesis of insular dysfunction. J Psychiatry Neurosci 37(1):17–27CrossRefPubMedPubMedCentral
go back to reference Palomero-Gallagher N, Vogt BA, Schleicher A, Mayberg HS, Zilles K (2009) Receptor architecture of human cingulate cortex: evaluation of the four-region neurobiological model. Hum Brain Mapp 30(8):2336–2355CrossRefPubMed Palomero-Gallagher N, Vogt BA, Schleicher A, Mayberg HS, Zilles K (2009) Receptor architecture of human cingulate cortex: evaluation of the four-region neurobiological model. Hum Brain Mapp 30(8):2336–2355CrossRefPubMed
go back to reference Parsey RV, Ojha A, Ogden RT, Erlandsson K, Kumar D, Landgrebe M, Van Heertum R, Mann JJ (2006) Metabolite considerations in the in vivo quantification of serotonin transporters using 11C-DASB and PET in humans. J Nucl Med 47(11):1796–1802PubMed Parsey RV, Ojha A, Ogden RT, Erlandsson K, Kumar D, Landgrebe M, Van Heertum R, Mann JJ (2006) Metabolite considerations in the in vivo quantification of serotonin transporters using 11C-DASB and PET in humans. J Nucl Med 47(11):1796–1802PubMed
go back to reference Pham TH, Mendez-David I, Defaix C, Guiard BP, Tritschler L, David DJ, Gardier AM (2016) Ketamine treatment involves medial prefrontal cortex serotonin to induce a rapid antidepressant-like activity in BALB/cJ mice. Neuropharmacology. doi:10.1016/j.neuropharm.2016.05.010 Pham TH, Mendez-David I, Defaix C, Guiard BP, Tritschler L, David DJ, Gardier AM (2016) Ketamine treatment involves medial prefrontal cortex serotonin to induce a rapid antidepressant-like activity in BALB/cJ mice. Neuropharmacology. doi:10.​1016/​j.​neuropharm.​2016.​05.​010
go back to reference Poels EM, Kegeles LS, Kantrowitz JT, Slifstein M, Javitt DC, Lieberman JA, Abi-Dargham A, Girgis RR (2014) Imaging glutamate in schizophrenia: review of findings and implications for drug discovery. Mol Psychiatry 19(1):20–29CrossRefPubMed Poels EM, Kegeles LS, Kantrowitz JT, Slifstein M, Javitt DC, Lieberman JA, Abi-Dargham A, Girgis RR (2014) Imaging glutamate in schizophrenia: review of findings and implications for drug discovery. Mol Psychiatry 19(1):20–29CrossRefPubMed
go back to reference Rowland LM, Bustillo JR, Mullins PG, Jung RE, Lenroot R, Landgraf E, Barrow R, Yeo R, Lauriello J, Brooks WM (2005) Effects of ketamine on anterior cingulate glutamate metabolism in healthy humans: a 4-T proton MRS study. Am J Psychiatry 162(2):394–396CrossRefPubMed Rowland LM, Bustillo JR, Mullins PG, Jung RE, Lenroot R, Landgraf E, Barrow R, Yeo R, Lauriello J, Brooks WM (2005) Effects of ketamine on anterior cingulate glutamate metabolism in healthy humans: a 4-T proton MRS study. Am J Psychiatry 162(2):394–396CrossRefPubMed
go back to reference Scheidegger M, Walter M, Lehmann M, Metzger C, Grimm S, Boeker H, Boesiger P, Henning A, Seifritz E (2012) Ketamine decreases resting state functional network connectivity in healthy subjects: implications for antidepressant drug action. PLoS One 7(9):e44799CrossRefPubMedPubMedCentral Scheidegger M, Walter M, Lehmann M, Metzger C, Grimm S, Boeker H, Boesiger P, Henning A, Seifritz E (2012) Ketamine decreases resting state functional network connectivity in healthy subjects: implications for antidepressant drug action. PLoS One 7(9):e44799CrossRefPubMedPubMedCentral
go back to reference Scheidegger M, Henning A, Walter M, Lehmann M, Kraehenmann R, Boeker H, Seifritz E, Grimm S (2016) Ketamine administration reduces amygdalo-hippocampal reactivity to emotional stimulation. Hum Brain Mapp 37(5):1941–1952CrossRefPubMed Scheidegger M, Henning A, Walter M, Lehmann M, Kraehenmann R, Boeker H, Seifritz E, Grimm S (2016) Ketamine administration reduces amygdalo-hippocampal reactivity to emotional stimulation. Hum Brain Mapp 37(5):1941–1952CrossRefPubMed
go back to reference Shcherbinin S, Doyle O, Zelaya FO, de Simoni S, Mehta MA, Schwarz AJ (2015) Modulatory effects of ketamine, risperidone and lamotrigine on resting brain perfusion in healthy human subjects. Psychopharmacology 232(21–22):4191–4204CrossRefPubMed Shcherbinin S, Doyle O, Zelaya FO, de Simoni S, Mehta MA, Schwarz AJ (2015) Modulatory effects of ketamine, risperidone and lamotrigine on resting brain perfusion in healthy human subjects. Psychopharmacology 232(21–22):4191–4204CrossRefPubMed
go back to reference Stone JM, Dietrich C, Edden R, Mehta MA, De Simoni S, Reed LJ, Krystal JH, Nutt D, Barker GJ (2012) Ketamine effects on brain GABA and glutamate levels with 1H-MRS: relationship to ketamine-induced psychopathology. Mol Psychiatry 17(7):664–665CrossRefPubMed Stone JM, Dietrich C, Edden R, Mehta MA, De Simoni S, Reed LJ, Krystal JH, Nutt D, Barker GJ (2012) Ketamine effects on brain GABA and glutamate levels with 1H-MRS: relationship to ketamine-induced psychopathology. Mol Psychiatry 17(7):664–665CrossRefPubMed
go back to reference Taylor KS, Seminowicz DA, Davis KD (2009) Two systems of resting state connectivity between the insula and cingulate cortex. Hum Brain Mapp 30(9):2731–2745CrossRefPubMed Taylor KS, Seminowicz DA, Davis KD (2009) Two systems of resting state connectivity between the insula and cingulate cortex. Hum Brain Mapp 30(9):2731–2745CrossRefPubMed
go back to reference Taylor MJ, Tiangga ER, Mhuircheartaigh RN, Cowen PJ (2012) Lack of effect of ketamine on cortical glutamate and glutamine in healthy volunteers: a proton magnetic resonance spectroscopy study. J Psychopharmacol 26(5):733–737CrossRefPubMedPubMedCentral Taylor MJ, Tiangga ER, Mhuircheartaigh RN, Cowen PJ (2012) Lack of effect of ketamine on cortical glutamate and glutamine in healthy volunteers: a proton magnetic resonance spectroscopy study. J Psychopharmacol 26(5):733–737CrossRefPubMedPubMedCentral
go back to reference Theberge J, Bartha R, Drost DJ, Menon RS, Malla A, Takhar J, Neufeld RW, Rogers J, Pavlosky W, Schaefer B, Densmore M, Al-Semaan Y, Williamson PC (2002) Glutamate and glutamine measured with 4.0 T proton MRS in never-treated patients with schizophrenia and healthy volunteers. Am J Psychiatry 159(11):1944–1946CrossRefPubMed Theberge J, Bartha R, Drost DJ, Menon RS, Malla A, Takhar J, Neufeld RW, Rogers J, Pavlosky W, Schaefer B, Densmore M, Al-Semaan Y, Williamson PC (2002) Glutamate and glutamine measured with 4.0 T proton MRS in never-treated patients with schizophrenia and healthy volunteers. Am J Psychiatry 159(11):1944–1946CrossRefPubMed
go back to reference Vollenweider FX, Vontobel P, Øye I, Hell D, Leenders KL (2000) Effects of (S)-ketamine on striatal dopamine: a [11C]raclopride PET study of a model psychosis in humans. J Psychiatr Res 34(1):35–43CrossRefPubMed Vollenweider FX, Vontobel P, Øye I, Hell D, Leenders KL (2000) Effects of (S)-ketamine on striatal dopamine: a [11C]raclopride PET study of a model psychosis in humans. J Psychiatr Res 34(1):35–43CrossRefPubMed
go back to reference Walter M, Li S, Demenescu LR (2014) Multistage drug effects of ketamine in the treatment of major depression. Eur Arch Psychiatry Clin Neurosci 264(Suppl 1):S55–S65CrossRefPubMed Walter M, Li S, Demenescu LR (2014) Multistage drug effects of ketamine in the treatment of major depression. Eur Arch Psychiatry Clin Neurosci 264(Suppl 1):S55–S65CrossRefPubMed
go back to reference Weissenbacher A, Kasess C, Gerstl F, Lanzenberger R, Moser E, Windischberger C (2009) Correlations and anticorrelations in resting-state functional connectivity MRI: a quantitative comparison of preprocessing strategies. Neuroimage 47(4):1408–1416CrossRefPubMed Weissenbacher A, Kasess C, Gerstl F, Lanzenberger R, Moser E, Windischberger C (2009) Correlations and anticorrelations in resting-state functional connectivity MRI: a quantitative comparison of preprocessing strategies. Neuroimage 47(4):1408–1416CrossRefPubMed
go back to reference Wise RG, Rogers R, Painter D, Bantick S, Ploghaus A, Williams P, Rapeport G, Tracey I (2002) Combining fMRI with a pharmacokinetic model to determine which brain areas activated by painful stimulation are specifically modulated by remifentanil. Neuroimage 16(4):999–1014CrossRefPubMed Wise RG, Rogers R, Painter D, Bantick S, Ploghaus A, Williams P, Rapeport G, Tracey I (2002) Combining fMRI with a pharmacokinetic model to determine which brain areas activated by painful stimulation are specifically modulated by remifentanil. Neuroimage 16(4):999–1014CrossRefPubMed
go back to reference Witkin JM, Monn JA, Schoepp DD, Li X, Overshiner C, Mitchell SN, Carter G, Johnson B, Rasmussen K, Rorick-Kehn LM (2016) The rapidly acting antidepressant ketamine and the mGlu2/3 receptor antagonist LY341495 rapidly engage dopaminergic mood circuits. J Pharmacol Exp Ther 358(1):71–82CrossRefPubMed Witkin JM, Monn JA, Schoepp DD, Li X, Overshiner C, Mitchell SN, Carter G, Johnson B, Rasmussen K, Rorick-Kehn LM (2016) The rapidly acting antidepressant ketamine and the mGlu2/3 receptor antagonist LY341495 rapidly engage dopaminergic mood circuits. J Pharmacol Exp Ther 358(1):71–82CrossRefPubMed
go back to reference Yu C, Zhou Y, Liu Y, Jiang T, Dong H, Zhang Y, Walter M (2011) Functional segregation of the human cingulate cortex is confirmed by functional connectivity based neuroanatomical parcellation. Neuroimage 54(4):2571–2581CrossRefPubMed Yu C, Zhou Y, Liu Y, Jiang T, Dong H, Zhang Y, Walter M (2011) Functional segregation of the human cingulate cortex is confirmed by functional connectivity based neuroanatomical parcellation. Neuroimage 54(4):2571–2581CrossRefPubMed
go back to reference Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, Charney DS, Manji HK (2006) A randomized trial of an N-methyl-d-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry 63(8):856–864CrossRefPubMed Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, Charney DS, Manji HK (2006) A randomized trial of an N-methyl-d-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry 63(8):856–864CrossRefPubMed
go back to reference Zhang WN, Chang SH, Guo LY, Zhang KL, Wang J (2013) The neural correlates of reward-related processing in major depressive disorder: a meta-analysis of functional magnetic resonance imaging studies. J Affect Disord 151(2):531–539CrossRefPubMed Zhang WN, Chang SH, Guo LY, Zhang KL, Wang J (2013) The neural correlates of reward-related processing in major depressive disorder: a meta-analysis of functional magnetic resonance imaging studies. J Affect Disord 151(2):531–539CrossRefPubMed
Metadata
Title
Ketamine-dependent neuronal activation in healthy volunteers
Authors
Anna Höflich
Andreas Hahn
Martin Küblböck
Georg S. Kranz
Thomas Vanicek
Sebastian Ganger
Marie Spies
Christian Windischberger
Siegfried Kasper
Dietmar Winkler
Rupert Lanzenberger
Publication date
01-04-2017
Publisher
Springer Berlin Heidelberg
Published in
Brain Structure and Function / Issue 3/2017
Print ISSN: 1863-2653
Electronic ISSN: 1863-2661
DOI
https://doi.org/10.1007/s00429-016-1291-0

Other articles of this Issue 3/2017

Brain Structure and Function 3/2017 Go to the issue