Skip to main content
Erschienen in: CNS Drugs 3/2018

01.03.2018 | Review Article

Convergent Mechanisms Underlying Rapid Antidepressant Action

verfasst von: Panos Zanos, Scott M. Thompson, Ronald S. Duman, Carlos A. Zarate Jr., Todd D. Gould

Erschienen in: CNS Drugs | Ausgabe 3/2018

Einloggen, um Zugang zu erhalten

Abstract

Traditional pharmacological treatments for depression have a delayed therapeutic onset, ranging from several weeks to months, and there is a high percentage of individuals who never respond to treatment. In contrast, ketamine produces rapid-onset antidepressant, anti-suicidal, and anti-anhedonic actions following a single administration to patients with depression. Proposed mechanisms of the antidepressant action of ketamine include N-methyl-d-aspartate receptor (NMDAR) modulation, gamma aminobutyric acid (GABA)-ergic interneuron disinhibition, and direct actions of its hydroxynorketamine (HNK) metabolites. Downstream actions include activation of the mechanistic target of rapamycin (mTOR), deactivation of glycogen synthase kinase-3 and eukaryotic elongation factor 2 (eEF2), enhanced brain-derived neurotrophic factor (BDNF) signaling, and activation of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors (AMPARs). These putative mechanisms of ketamine action are not mutually exclusive and may complement each other to induce potentiation of excitatory synapses in affective-regulating brain circuits, which results in amelioration of depression symptoms. We review these proposed mechanisms of ketamine action in the context of how such mechanisms are informing the development of novel putative rapid-acting antidepressant drugs. Such drugs that have undergone pre-clinical, and in some cases clinical, testing include the muscarinic acetylcholine receptor antagonist scopolamine, GluN2B-NMDAR antagonists (i.e., CP-101,606, MK-0657), (2R,6R)-HNK, NMDAR glycine site modulators (i.e., 4-chlorokynurenine, pro-drug of the glycineB NMDAR antagonist 7-chlorokynurenic acid), NMDAR agonists [i.e., GLYX-13 (rapastinel)], metabotropic glutamate receptor 2/3 (mGluR2/3) antagonists, GABAA receptor modulators, and drugs acting on various serotonin receptor subtypes. These ongoing studies suggest that the future acute treatment of depression will typically occur within hours, rather than months, of treatment initiation.
Literatur
1.
Zurück zum Zitat 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(23):3095–105.PubMedCrossRef 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(23):3095–105.PubMedCrossRef
2.
Zurück zum Zitat Insel TR, Wang PS. The STAR*D trial: revealing the need for better treatments. Psychiatr Serv. 2009;60(11):1466–7.PubMedCrossRef Insel TR, Wang PS. The STAR*D trial: revealing the need for better treatments. Psychiatr Serv. 2009;60(11):1466–7.PubMedCrossRef
3.
Zurück zum Zitat Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Pychiatry. 2006;163(11):1905–17.CrossRef Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Pychiatry. 2006;163(11):1905–17.CrossRef
4.
Zurück zum Zitat Gelenberg AJ, Chesen CL. How fast are antidepressants? J Clin Psychiatry. 2000;61(10):712–21.PubMedCrossRef Gelenberg AJ, Chesen CL. How fast are antidepressants? J Clin Psychiatry. 2000;61(10):712–21.PubMedCrossRef
5.
Zurück zum Zitat Insel TR, Scolnick EM. Cure therapeutics and strategic prevention: raising the bar for mental health research. Mol Psychiatry. 2006;11(1):11–7.PubMedPubMedCentralCrossRef Insel TR, Scolnick EM. Cure therapeutics and strategic prevention: raising the bar for mental health research. Mol Psychiatry. 2006;11(1):11–7.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Short B, Fong J, Galvez V, Shelker W, Loo CK. Side-effects associated with ketamine use in depression: a systematic review. Lancet Psychiatry. 2018;5(1):65–78.PubMedCrossRef Short B, Fong J, Galvez V, Shelker W, Loo CK. Side-effects associated with ketamine use in depression: a systematic review. Lancet Psychiatry. 2018;5(1):65–78.PubMedCrossRef
7.
Zurück zum Zitat Segman RH, Shapira B, Gorfine M, Lerer B. Onset and time course of antidepressant action: psychopharmacological implications of a controlled trial of electroconvulsive therapy. Psychopharmacology (Berl). 1995;119(4):440–8.PubMedCrossRef Segman RH, Shapira B, Gorfine M, Lerer B. Onset and time course of antidepressant action: psychopharmacological implications of a controlled trial of electroconvulsive therapy. Psychopharmacology (Berl). 1995;119(4):440–8.PubMedCrossRef
8.
Zurück zum Zitat Husain MM, Rush AJ, Fink M, Knapp R, Petrides G, Rummans T, et al. Speed of response and remission in major depressive disorder with acute electroconvulsive therapy (ECT): a Consortium for Research in ECT (CORE) report. J Clin Psychiatry. 2004;65(4):485–91.PubMedCrossRef Husain MM, Rush AJ, Fink M, Knapp R, Petrides G, Rummans T, et al. Speed of response and remission in major depressive disorder with acute electroconvulsive therapy (ECT): a Consortium for Research in ECT (CORE) report. J Clin Psychiatry. 2004;65(4):485–91.PubMedCrossRef
9.
Zurück zum Zitat Kellner CH, Knapp RG, Petrides G, Rummans TA, Husain MM, Rasmussen K, et al. Continuation electroconvulsive therapy vs pharmacotherapy for relapse prevention in major depression: a multisite study from the Consortium for Research in Electroconvulsive Therapy (CORE). Arch Gen Psychiatry. 2006;63(12):1337–44.PubMedPubMedCentralCrossRef Kellner CH, Knapp RG, Petrides G, Rummans TA, Husain MM, Rasmussen K, et al. Continuation electroconvulsive therapy vs pharmacotherapy for relapse prevention in major depression: a multisite study from the Consortium for Research in Electroconvulsive Therapy (CORE). Arch Gen Psychiatry. 2006;63(12):1337–44.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Dierckx B, Heijnen WT, van den Broek WW, Birkenhager TK. Efficacy of electroconvulsive therapy in bipolar versus unipolar major depression: a meta-analysis. Bipolar Disord. 2012;14(2):146–50.PubMedCrossRef Dierckx B, Heijnen WT, van den Broek WW, Birkenhager TK. Efficacy of electroconvulsive therapy in bipolar versus unipolar major depression: a meta-analysis. Bipolar Disord. 2012;14(2):146–50.PubMedCrossRef
11.
Zurück zum Zitat Post RM, Uhde TW, Rubinow DR, Huggins T. Differential time course of antidepressant effects after sleep deprivation, ECT, and carbamazepine: clinical and theoretical implications. Psychiatry Res. 1987;22(1):11–9.PubMedCrossRef Post RM, Uhde TW, Rubinow DR, Huggins T. Differential time course of antidepressant effects after sleep deprivation, ECT, and carbamazepine: clinical and theoretical implications. Psychiatry Res. 1987;22(1):11–9.PubMedCrossRef
12.
Zurück zum Zitat Nutt DJ, Gleiter CH, Glue P. Neuropharmacological aspects of ECT: in search of the primary mechanism of action. Convuls Ther. 1989;5(3):250–60.PubMed Nutt DJ, Gleiter CH, Glue P. Neuropharmacological aspects of ECT: in search of the primary mechanism of action. Convuls Ther. 1989;5(3):250–60.PubMed
13.
Zurück zum Zitat Nobler MS, Sackeim HA, Moeller JR, Prudic J, Petkova E, Waternaux C. Quantifying the speed of symptomatic improvement with electroconvulsive therapy: comparison of alternative statistical methods. Convuls Ther. 1997;13(4):208–21.PubMed Nobler MS, Sackeim HA, Moeller JR, Prudic J, Petkova E, Waternaux C. Quantifying the speed of symptomatic improvement with electroconvulsive therapy: comparison of alternative statistical methods. Convuls Ther. 1997;13(4):208–21.PubMed
14.
Zurück zum Zitat Houck W, Abonour R, Vance G, Einhorn LH. Secondary leukemias in refractory germ cell tumor patients undergoing autologous stem-cell transplantation using high-dose etoposide. J Clin Oncol. 2004;22(11):2155–8.PubMedCrossRef Houck W, Abonour R, Vance G, Einhorn LH. Secondary leukemias in refractory germ cell tumor patients undergoing autologous stem-cell transplantation using high-dose etoposide. J Clin Oncol. 2004;22(11):2155–8.PubMedCrossRef
15.
Zurück zum Zitat Wu JC, Bunney WE. The biological basis of an antidepressant response to sleep deprivation and relapse: review and hypothesis. Am J Psychiatry. 1990;147(1):14–21.PubMedCrossRef Wu JC, Bunney WE. The biological basis of an antidepressant response to sleep deprivation and relapse: review and hypothesis. Am J Psychiatry. 1990;147(1):14–21.PubMedCrossRef
16.
Zurück zum Zitat Wiegand M, Riemann D, Schreiber W, Lauer CJ, Berger M. Effect of morning and afternoon naps on mood after total sleep deprivation in patients with major depression. Biol Psychiatry. 1993;33(6):467–76.PubMedCrossRef Wiegand M, Riemann D, Schreiber W, Lauer CJ, Berger M. Effect of morning and afternoon naps on mood after total sleep deprivation in patients with major depression. Biol Psychiatry. 1993;33(6):467–76.PubMedCrossRef
18.
19.
Zurück zum Zitat Mion G. History of anaesthesia: the ketamine story: past, present and future. Eur J Anaesthesiol. 2017;34(9):571–5.PubMedCrossRef Mion G. History of anaesthesia: the ketamine story: past, present and future. Eur J Anaesthesiol. 2017;34(9):571–5.PubMedCrossRef
20.
Zurück zum Zitat 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(4):351–4.PubMedCrossRef 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(4):351–4.PubMedCrossRef
21.
Zurück zum Zitat 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(8):856–64.PubMedCrossRef 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(8):856–64.PubMedCrossRef
22.
Zurück zum Zitat Price RB, Iosifescu DV, Murrough JW, Chang LC, Al Jurdi RK, Iqbal SZ, et al. Effects of ketamine on explicit and implicit suicidal cognition: a randomized controlled trial in treatment-resistant depression. Depress Anxiety. 2014;31(4):335–43.PubMedPubMedCentralCrossRef Price RB, Iosifescu DV, Murrough JW, Chang LC, Al Jurdi RK, Iqbal SZ, et al. Effects of ketamine on explicit and implicit suicidal cognition: a randomized controlled trial in treatment-resistant depression. Depress Anxiety. 2014;31(4):335–43.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat 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;71(12):1605–11.PubMedPubMedCentralCrossRef 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;71(12):1605–11.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Lapidus KA, Levitch CF, Perez AM, Brallier JW, Parides MK, Soleimani L, et al. A randomized controlled trial of intranasal ketamine in major depressive disorder. Biol Psychiatry. 2014;76(12):970–6.PubMedPubMedCentralCrossRef Lapidus KA, Levitch CF, Perez AM, Brallier JW, Parides MK, Soleimani L, et al. A randomized controlled trial of intranasal ketamine in major depressive disorder. Biol Psychiatry. 2014;76(12):970–6.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Newport DJ, Carpenter LL, McDonald WM, Potash JB, Tohen M, Nemeroff CB. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172(10):950–66.PubMedCrossRef Newport DJ, Carpenter LL, McDonald WM, Potash JB, Tohen M, Nemeroff CB. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172(10):950–66.PubMedCrossRef
26.
Zurück zum Zitat McGirr A, Berlim MT, Bond DJ, Fleck MP, Yatham LN, Lam RW. A systematic review and meta-analysis of randomized, double-blind, placebo-controlled trials of ketamine in the rapid treatment of major depressive episodes. Psychol Med. 2015;45(4):693–704.PubMedCrossRef McGirr A, Berlim MT, Bond DJ, Fleck MP, Yatham LN, Lam RW. A systematic review and meta-analysis of randomized, double-blind, placebo-controlled trials of ketamine in the rapid treatment of major depressive episodes. Psychol Med. 2015;45(4):693–704.PubMedCrossRef
27.
Zurück zum Zitat Romeo B, Choucha W, Fossati P, Rotge JY. Meta-analysis of short- and mid-term efficacy of ketamine in unipolar and bipolar depression. Psychiatry Res. 2015;230(2):682–8.PubMedCrossRef Romeo B, Choucha W, Fossati P, Rotge JY. Meta-analysis of short- and mid-term efficacy of ketamine in unipolar and bipolar depression. Psychiatry Res. 2015;230(2):682–8.PubMedCrossRef
28.
Zurück zum Zitat Kishimoto T, Chawla JM, Hagi K, Zarate CA, Kane JM, Bauer M, et al. Single-dose infusion ketamine and non-ketamine N-methyl-d-aspartate receptor antagonists for unipolar and bipolar depression: a meta-analysis of efficacy, safety and time trajectories. Psychol Med. 2016;46(7):1459–72.PubMedPubMedCentralCrossRef Kishimoto T, Chawla JM, Hagi K, Zarate CA, Kane JM, Bauer M, et al. Single-dose infusion ketamine and non-ketamine N-methyl-d-aspartate receptor antagonists for unipolar and bipolar depression: a meta-analysis of efficacy, safety and time trajectories. Psychol Med. 2016;46(7):1459–72.PubMedPubMedCentralCrossRef
29.
30.
Zurück zum Zitat 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;51(3):199–214.PubMedCrossRef 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;51(3):199–214.PubMedCrossRef
31.
Zurück zum Zitat Sanacora G, Frye MA, McDonald W, Mathew SJ, Turner MS, Schatzberg AF, et al. A consensus statement on the use of ketamine in the treatment of mood disorders. JAMA Psychiatry. 2017;74(4):399–405.PubMedCrossRef Sanacora G, Frye MA, McDonald W, Mathew SJ, Turner MS, Schatzberg AF, et al. A consensus statement on the use of ketamine in the treatment of mood disorders. JAMA Psychiatry. 2017;74(4):399–405.PubMedCrossRef
32.
Zurück zum Zitat Ramaker MJ, Dulawa SC. Identifying fast-onset antidepressants using rodent models. Mol Psychiatry. 2017;22(5):656–65.PubMedCrossRef Ramaker MJ, Dulawa SC. Identifying fast-onset antidepressants using rodent models. Mol Psychiatry. 2017;22(5):656–65.PubMedCrossRef
33.
Zurück zum Zitat Mion G, Villevieille T. Ketamine pharmacology: an update (pharmacodynamics and molecular aspects, recent findings). CNS Neurosci Ther. 2013;19(6):370–80.PubMedCrossRef Mion G, Villevieille T. Ketamine pharmacology: an update (pharmacodynamics and molecular aspects, recent findings). CNS Neurosci Ther. 2013;19(6):370–80.PubMedCrossRef
34.
Zurück zum Zitat Kohrs R, Durieux ME. Ketamine: teaching an old drug new tricks. Anesth Analg. 1998;87(5):1186–93.PubMed Kohrs R, Durieux ME. Ketamine: teaching an old drug new tricks. Anesth Analg. 1998;87(5):1186–93.PubMed
35.
Zurück zum Zitat Singh JB, Fedgchin M, Daly E, Xi L, Melman C, De Bruecker G, et al. Intravenous esketamine in adult treatment-resistant depression: a double-blind, double-randomization, placebo-controlled study. Biol Psychiatry. 2016;80(6):424–31.PubMedCrossRef Singh JB, Fedgchin M, Daly E, Xi L, Melman C, De Bruecker G, et al. Intravenous esketamine in adult treatment-resistant depression: a double-blind, double-randomization, placebo-controlled study. Biol Psychiatry. 2016;80(6):424–31.PubMedCrossRef
36.
Zurück zum Zitat 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;170(10):1134–42.PubMedPubMedCentralCrossRef 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;170(10):1134–42.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Diazgranados N, Ibrahim L, Brutsche NE, Newberg A, Kronstein P, Khalife S, et al. A randomized add-on trial of an N-methyl-d-aspartate antagonist in treatment-resistant bipolar depression. Arch Gen Psychiatry. 2010;67(8):793–802.PubMedPubMedCentralCrossRef Diazgranados N, Ibrahim L, Brutsche NE, Newberg A, Kronstein P, Khalife S, et al. A randomized add-on trial of an N-methyl-d-aspartate antagonist in treatment-resistant bipolar depression. Arch Gen Psychiatry. 2010;67(8):793–802.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat 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;71(11):939–46.PubMedPubMedCentralCrossRef 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;71(11):939–46.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Liu R-J, Fuchikami M, Dwyer JM, Lepack AE, Duman RS, Aghajanian GK. GSK-3 inhibition potentiates the synaptogenic and antidepressant-like effects of subthreshold doses of ketamine. Neuropsychopharmacology. 2013;38(11):2268–77.PubMedPubMedCentralCrossRef Liu R-J, Fuchikami M, Dwyer JM, Lepack AE, Duman RS, Aghajanian GK. GSK-3 inhibition potentiates the synaptogenic and antidepressant-like effects of subthreshold doses of ketamine. Neuropsychopharmacology. 2013;38(11):2268–77.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Lally N, Nugent AC, Luckenbaugh DA, Ameli R, Roiser JP, Zarate CA. Anti-anhedonic effect of ketamine and its neural correlates in treatment-resistant bipolar depression. Transl Psychiatry. 2014;4(10):e469.PubMedPubMedCentralCrossRef Lally N, Nugent AC, Luckenbaugh DA, Ameli R, Roiser JP, Zarate CA. Anti-anhedonic effect of ketamine and its neural correlates in treatment-resistant bipolar depression. Transl Psychiatry. 2014;4(10):e469.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Ballard ED, Ionescu DF, Vande Voort JL, Niciu MJ, Richards EM, Luckenbaugh DA, et al. Improvement in suicidal ideation after ketamine infusion: relationship to reductions in depression and anxiety. J Psychiatr Res. 2014;58:161–6.PubMedCrossRef Ballard ED, Ionescu DF, Vande Voort JL, Niciu MJ, Richards EM, Luckenbaugh DA, et al. Improvement in suicidal ideation after ketamine infusion: relationship to reductions in depression and anxiety. J Psychiatr Res. 2014;58:161–6.PubMedCrossRef
42.
Zurück zum Zitat 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(5994):959–64.PubMedPubMedCentralCrossRef 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(5994):959–64.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat 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;475(7354):91–5.PubMedPubMedCentralCrossRef 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;475(7354):91–5.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Zanos P, Moaddel R, Morris PJ, Georgiou P, Fischell J, Elmer GI, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016;533(7604):481–6.PubMedPubMedCentralCrossRef Zanos P, Moaddel R, Morris PJ, Georgiou P, Fischell J, Elmer GI, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016;533(7604):481–6.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Zanos P, Bhat S, Terrillion CE, Smith RJ, Tonelli LH, Gould TD. Sex-dependent modulation of age-related cognitive decline by the L-type calcium channel gene Cacna1c (Cav 1.2). Eur J Neurosci. 2015;42(8):2499–507.PubMedPubMedCentralCrossRef Zanos P, Bhat S, Terrillion CE, Smith RJ, Tonelli LH, Gould TD. Sex-dependent modulation of age-related cognitive decline by the L-type calcium channel gene Cacna1c (Cav 1.2). Eur J Neurosci. 2015;42(8):2499–507.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat 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;63(4):349–52.PubMedCrossRef 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;63(4):349–52.PubMedCrossRef
48.
Zurück zum Zitat Belujon P, Grace AA. Restoring mood balance in depression: ketamine reverses deficit in dopamine-dependent synaptic plasticity. Biol Psychiatry. 2014;76(12):927–36.PubMedPubMedCentralCrossRef Belujon P, Grace AA. Restoring mood balance in depression: ketamine reverses deficit in dopamine-dependent synaptic plasticity. Biol Psychiatry. 2014;76(12):927–36.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Koike H, Iijima M, Chaki S. Involvement of the mammalian target of rapamycin signaling in the antidepressant-like effect of group II metabotropic glutamate receptor antagonists. Neuropharmacology. 2011;61(8):1419–23.PubMedCrossRef Koike H, Iijima M, Chaki S. Involvement of the mammalian target of rapamycin signaling in the antidepressant-like effect of group II metabotropic glutamate receptor antagonists. Neuropharmacology. 2011;61(8):1419–23.PubMedCrossRef
50.
Zurück zum Zitat Zhang H, Xue W, Wu R, Gong T, Tao W, Zhou X, et al. Rapid antidepressant activity of ethanol extract of Gardenia jasminoides Ellis is associated with upregulation of BDNF expression in the hippocampus. Evid Based Complement Alternat Med. 2015;2015:761238.PubMedPubMedCentral Zhang H, Xue W, Wu R, Gong T, Tao W, Zhou X, et al. Rapid antidepressant activity of ethanol extract of Gardenia jasminoides Ellis is associated with upregulation of BDNF expression in the hippocampus. Evid Based Complement Alternat Med. 2015;2015:761238.PubMedPubMedCentral
51.
Zurück zum Zitat Beurel E, Song L, Jope RS. Inhibition of glycogen synthase kinase-3 is necessary for the rapid antidepressant effect of ketamine in mice. Mol Psychiatry. 2011;16(11):1068–70.PubMedPubMedCentralCrossRef Beurel E, Song L, Jope RS. Inhibition of glycogen synthase kinase-3 is necessary for the rapid antidepressant effect of ketamine in mice. Mol Psychiatry. 2011;16(11):1068–70.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Carrier N, Kabbaj M. Sex differences in the antidepressant-like effects of ketamine. Neuropharmacology. 2013;70:27–34.PubMedCrossRef Carrier N, Kabbaj M. Sex differences in the antidepressant-like effects of ketamine. Neuropharmacology. 2013;70:27–34.PubMedCrossRef
53.
Zurück zum Zitat Gideons ES, Kavalali ET, Monteggia LM. Mechanisms underlying differential effectiveness of memantine and ketamine in rapid antidepressant responses. Proc Natl Acad Sci USA. 2014;111(23):8649–54.PubMedPubMedCentralCrossRef Gideons ES, Kavalali ET, Monteggia LM. Mechanisms underlying differential effectiveness of memantine and ketamine in rapid antidepressant responses. Proc Natl Acad Sci USA. 2014;111(23):8649–54.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Iijima M, Fukumoto K, Chaki S. Acute and sustained effects of a metabotropic glutamate 5 receptor antagonist in the novelty-suppressed feeding test. Behav Brain Res. 2012;235(2):287–92.PubMedCrossRef Iijima M, Fukumoto K, Chaki S. Acute and sustained effects of a metabotropic glutamate 5 receptor antagonist in the novelty-suppressed feeding test. Behav Brain Res. 2012;235(2):287–92.PubMedCrossRef
55.
Zurück zum Zitat 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(8):754–61.PubMedPubMedCentralCrossRef 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(8):754–61.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Louderback KM, Wills TA, Muglia LJ, Winder DG. Knockdown of BNST GluN2B-containing NMDA receptors mimics the actions of ketamine on novelty-induced hypophagia. Transl Psychiatry. 2013;3(3):e331.PubMedPubMedCentralCrossRef Louderback KM, Wills TA, Muglia LJ, Winder DG. Knockdown of BNST GluN2B-containing NMDA receptors mimics the actions of ketamine on novelty-induced hypophagia. Transl Psychiatry. 2013;3(3):e331.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Burgdorf J, Zhang XL, Nicholson KL, Balster RL, Leander JD, Stanton PK, et al. GLYX-13, a NMDA receptor glycine-site functional partial agonist, induces antidepressant-like effects without ketamine-like side effects. Neuropsychopharmacology. 2013;38(5):729–42.PubMedPubMedCentralCrossRef Burgdorf J, Zhang XL, Nicholson KL, Balster RL, Leander JD, Stanton PK, et al. GLYX-13, a NMDA receptor glycine-site functional partial agonist, induces antidepressant-like effects without ketamine-like side effects. Neuropsychopharmacology. 2013;38(5):729–42.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Yang B, Zhang JC, Han M, Yao W, Yang C, Ren Q, et al. Comparison of R-ketamine and rapastinel antidepressant effects in the social defeat stress model of depression. Psychopharmacology (Berl). 2016;233(19–20):3647–57.PubMedPubMedCentralCrossRef Yang B, Zhang JC, Han M, Yao W, Yang C, Ren Q, et al. Comparison of R-ketamine and rapastinel antidepressant effects in the social defeat stress model of depression. Psychopharmacology (Berl). 2016;233(19–20):3647–57.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Donahue RJ, Muschamp JW, Russo SJ, Nestler EJ, Carlezon WA Jr. Effects of striatal DeltaFosB overexpression and ketamine on social defeat stress-induced anhedonia in mice. Biol Psychiatry. 2014;76(7):550–8.PubMedPubMedCentralCrossRef Donahue RJ, Muschamp JW, Russo SJ, Nestler EJ, Carlezon WA Jr. Effects of striatal DeltaFosB overexpression and ketamine on social defeat stress-induced anhedonia in mice. Biol Psychiatry. 2014;76(7):550–8.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Zhang JC, Yao W, Dong C, Yang C, Ren Q, Ma M, et al. Comparison of ketamine, 7,8-dihydroxyflavone, and ANA-12 antidepressant effects in the social defeat stress model of depression. Psychopharmacology (Berl). 2015;232(23):4325–35.PubMedCrossRef Zhang JC, Yao W, Dong C, Yang C, Ren Q, Ma M, et al. Comparison of ketamine, 7,8-dihydroxyflavone, and ANA-12 antidepressant effects in the social defeat stress model of depression. Psychopharmacology (Berl). 2015;232(23):4325–35.PubMedCrossRef
61.
Zurück zum Zitat 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(9):776–86.PubMedCrossRef 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(9):776–86.PubMedCrossRef
62.
Zurück zum Zitat Gould TD, Georgiou P, Brenner LA, Brundin L, Can A, Courtet P, et al. Animal models to improve our understanding and treatment of suicidal behavior. Transl Psychiatry. 2017;7(4):e1092.PubMedPubMedCentralCrossRef Gould TD, Georgiou P, Brenner LA, Brundin L, Can A, Courtet P, et al. Animal models to improve our understanding and treatment of suicidal behavior. Transl Psychiatry. 2017;7(4):e1092.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Daly EJ, Singh JB, Fedgchin M, Cooper K, Lim P, Shelton RC, et al. Efficacy and safety of intranasal esketamine adjunctive to oral antidepressant therapy in treatment-resistant depression: A randomized clinical trial. JAMA Psychiatry. 2018;75(2):139–48.PubMedCrossRef Daly EJ, Singh JB, Fedgchin M, Cooper K, Lim P, Shelton RC, et al. Efficacy and safety of intranasal esketamine adjunctive to oral antidepressant therapy in treatment-resistant depression: A randomized clinical trial. JAMA Psychiatry. 2018;75(2):139–48.PubMedCrossRef
64.
Zurück zum Zitat Yang C, Shirayama Y, Zhang JC, Ren Q, Yao W, Ma M, et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry. 2015;5:e632.PubMedPubMedCentralCrossRef Yang C, Shirayama Y, Zhang JC, Ren Q, Yao W, Ma M, et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry. 2015;5:e632.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Fukumoto K, Toki H, Iijima M, Hashihayata T, J-i Yamaguchi, Hashimoto K, et al. Antidepressant potential of (R)-ketamine in rodent models: comparison with (S)-ketamine. J Pharmacol Exp Ther. 2017;361(1):9–16.PubMedCrossRef Fukumoto K, Toki H, Iijima M, Hashihayata T, J-i Yamaguchi, Hashimoto K, et al. Antidepressant potential of (R)-ketamine in rodent models: comparison with (S)-ketamine. J Pharmacol Exp Ther. 2017;361(1):9–16.PubMedCrossRef
66.
67.
Zurück zum Zitat Morgan CJ, Mofeez A, Brandner B, Bromley L, Curran HV. Ketamine impairs response inhibition and is positively reinforcing in healthy volunteers: a dose-response study. Psychopharmacology (Berl). 2004;172(3):298–308.PubMed Morgan CJ, Mofeez A, Brandner B, Bromley L, Curran HV. Ketamine impairs response inhibition and is positively reinforcing in healthy volunteers: a dose-response study. Psychopharmacology (Berl). 2004;172(3):298–308.PubMed
68.
Zurück zum Zitat Malhotra AK, Pinals DA, Weingartner H, Sirocco K, Missar CD, Pickar D, et al. NMDA receptor function and human cognition: the effects of ketamine in healthy volunteers. Neuropsychopharmacology. 1996;14(5):301–7.PubMedCrossRef Malhotra AK, Pinals DA, Weingartner H, Sirocco K, Missar CD, Pickar D, et al. NMDA receptor function and human cognition: the effects of ketamine in healthy volunteers. Neuropsychopharmacology. 1996;14(5):301–7.PubMedCrossRef
69.
Zurück zum Zitat Curran HV, Monaghan L. In and out of the K-hole: a comparison of the acute and residual effects of ketamine in frequent and infrequent ketamine users. Addiction. 2001;96(5):749–60.PubMedCrossRef Curran HV, Monaghan L. In and out of the K-hole: a comparison of the acute and residual effects of ketamine in frequent and infrequent ketamine users. Addiction. 2001;96(5):749–60.PubMedCrossRef
70.
71.
Zurück zum Zitat Burke RE. The relative selectivity of anticholinergic drugs for the M1 and M2 muscarinic receptor subtypes. Mov Disord. 1986;1(2):135–44.PubMedCrossRef Burke RE. The relative selectivity of anticholinergic drugs for the M1 and M2 muscarinic receptor subtypes. Mov Disord. 1986;1(2):135–44.PubMedCrossRef
72.
Zurück zum Zitat Browne RG. Effects of antidepressants and anticholinergics in a mouse “behavioral despair” test. Eur J Pharmacol. 1979;58(3):331–4.PubMedCrossRef Browne RG. Effects of antidepressants and anticholinergics in a mouse “behavioral despair” test. Eur J Pharmacol. 1979;58(3):331–4.PubMedCrossRef
73.
Zurück zum Zitat Betin C, DeFeudis FV, Blavet N, Clostre F. Further characterization of the behavioral despair test in mice: positive effects of convulsants. Physiol Behav. 1982;28(2):307–11.PubMedCrossRef Betin C, DeFeudis FV, Blavet N, Clostre F. Further characterization of the behavioral despair test in mice: positive effects of convulsants. Physiol Behav. 1982;28(2):307–11.PubMedCrossRef
74.
Zurück zum Zitat Kasper S, Moises HW, Beckmann H. The anticholinergic biperiden in depressive disorders. Pharmacopsychiatria. 1981;14(6):195–8.PubMedCrossRef Kasper S, Moises HW, Beckmann H. The anticholinergic biperiden in depressive disorders. Pharmacopsychiatria. 1981;14(6):195–8.PubMedCrossRef
75.
Zurück zum Zitat Gillin JC, Sutton L, Ruiz C, Darko D, Golshan S, Risch SC, et al. The effects of scopolamine on sleep and mood in depressed patients with a history of alcoholism and a normal comparison group. Biol Psychiatry. 1991;30(2):157–69.PubMedCrossRef Gillin JC, Sutton L, Ruiz C, Darko D, Golshan S, Risch SC, et al. The effects of scopolamine on sleep and mood in depressed patients with a history of alcoholism and a normal comparison group. Biol Psychiatry. 1991;30(2):157–69.PubMedCrossRef
76.
Zurück zum Zitat Furey ML, Drevets WC. Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled clinical trial. Arch Gen Psychiatry. 2006;63(10):1121–9.PubMedPubMedCentralCrossRef Furey ML, Drevets WC. Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled clinical trial. Arch Gen Psychiatry. 2006;63(10):1121–9.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Drevets WC, Furey ML. Replication of scopolamine’s antidepressant efficacy in major depressive disorder: a randomized, placebo-controlled clinical trial. Biol Psychiatry. 2010;67(5):432–8.PubMedPubMedCentralCrossRef Drevets WC, Furey ML. Replication of scopolamine’s antidepressant efficacy in major depressive disorder: a randomized, placebo-controlled clinical trial. Biol Psychiatry. 2010;67(5):432–8.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Furey ML, Khanna A, Hoffman EM, Drevets WC. Scopolamine produces larger antidepressant and antianxiety effects in women than in men. Neuropsychopharmacology. 2010;35(12):2479–88.PubMedPubMedCentralCrossRef Furey ML, Khanna A, Hoffman EM, Drevets WC. Scopolamine produces larger antidepressant and antianxiety effects in women than in men. Neuropsychopharmacology. 2010;35(12):2479–88.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Howland RH. The antidepressant effects of anticholinergic drugs. J Psychosoc Nurs Ment Health Serv. 2009;47(6):17–20.CrossRef Howland RH. The antidepressant effects of anticholinergic drugs. J Psychosoc Nurs Ment Health Serv. 2009;47(6):17–20.CrossRef
80.
Zurück zum Zitat Gillin JC, Lauriello J, Kelsoe JR, Rapaport M, Golshan S, Kenny WM, et al. No antidepressant effect of biperiden compared with placebo in depression: a double-blind 6-week clinical trial. Psychiatry Res. 1995;58(2):99–105.PubMedCrossRef Gillin JC, Lauriello J, Kelsoe JR, Rapaport M, Golshan S, Kenny WM, et al. No antidepressant effect of biperiden compared with placebo in depression: a double-blind 6-week clinical trial. Psychiatry Res. 1995;58(2):99–105.PubMedCrossRef
81.
Zurück zum Zitat Navarria A, Wohleb ES, Voleti B, Ota KT, Dutheil S, Lepack AE, et al. Rapid antidepressant actions of scopolamine: Role of medial prefrontal cortex and M1-subtype muscarinic acetylcholine receptors. Neurobiol Dis. 2015;82:254–61.PubMedPubMedCentralCrossRef Navarria A, Wohleb ES, Voleti B, Ota KT, Dutheil S, Lepack AE, et al. Rapid antidepressant actions of scopolamine: Role of medial prefrontal cortex and M1-subtype muscarinic acetylcholine receptors. Neurobiol Dis. 2015;82:254–61.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Wohleb ES, Wu M, Gerhard DM, Taylor SR, Picciotto MR, Alreja M, et al. GABA interneurons mediate the rapid antidepressant-like effects of scopolamine. J Clin Invest. 2016;126(7):2482–94.PubMedPubMedCentralCrossRef Wohleb ES, Wu M, Gerhard DM, Taylor SR, Picciotto MR, Alreja M, et al. GABA interneurons mediate the rapid antidepressant-like effects of scopolamine. J Clin Invest. 2016;126(7):2482–94.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Geoffroy M, Scheel-Kruger J, Christensen AV. Effect of imipramine in the “learned helplessness” model of depression in rats is not mimicked by combinations of specific reuptake inhibitors and scopolamine. Psychopharmacology (Berl). 1990;101(3):371–5.PubMedCrossRef Geoffroy M, Scheel-Kruger J, Christensen AV. Effect of imipramine in the “learned helplessness” model of depression in rats is not mimicked by combinations of specific reuptake inhibitors and scopolamine. Psychopharmacology (Berl). 1990;101(3):371–5.PubMedCrossRef
84.
Zurück zum Zitat Anisman H, Remington G, Sklar LS. Effect of inescapable shock on subsequent escape performance: catecholaminergic and cholinergic mediation of response initiation and maintenance. Psychopharmacology (Berl). 1979;61(2):107–24.PubMedCrossRef Anisman H, Remington G, Sklar LS. Effect of inescapable shock on subsequent escape performance: catecholaminergic and cholinergic mediation of response initiation and maintenance. Psychopharmacology (Berl). 1979;61(2):107–24.PubMedCrossRef
85.
Zurück zum Zitat 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(10):742–9.PubMedCrossRef 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(10):742–9.PubMedCrossRef
86.
Zurück zum Zitat Witkin JM, Overshiner C, Li X, Catlow JT, Wishart GN, Schober DA, et al. M1 and m2 muscarinic receptor subtypes regulate antidepressant-like effects of the rapidly acting antidepressant scopolamine. J Pharmacol Exp Ther. 2014;351(2):448–56.PubMedCrossRef Witkin JM, Overshiner C, Li X, Catlow JT, Wishart GN, Schober DA, et al. M1 and m2 muscarinic receptor subtypes regulate antidepressant-like effects of the rapidly acting antidepressant scopolamine. J Pharmacol Exp Ther. 2014;351(2):448–56.PubMedCrossRef
87.
88.
Zurück zum Zitat Vyklicky V, Korinek M, Smejkalova T, Balik A, Krausova B, Kaniakova M, et al. Structure, function, and pharmacology of NMDA receptor channels. Physiol Res. 2014;63(Suppl. 1):S191–203.PubMed Vyklicky V, Korinek M, Smejkalova T, Balik A, Krausova B, Kaniakova M, et al. Structure, function, and pharmacology of NMDA receptor channels. Physiol Res. 2014;63(Suppl. 1):S191–203.PubMed
89.
Zurück zum Zitat Trullas R, Skolnick P. Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. Eur J Pharmacol. 1990;185(1):1–10.PubMedCrossRef Trullas R, Skolnick P. Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. Eur J Pharmacol. 1990;185(1):1–10.PubMedCrossRef
90.
Zurück zum Zitat Paul IA, Nowak G, Layer RT, Popik P, Skolnick P. Adaptation of the N-methyl-d-aspartate receptor complex following chronic antidepressant treatments. J Pharmacol Exp Ther. 1994;269(1):95–102.PubMed Paul IA, Nowak G, Layer RT, Popik P, Skolnick P. Adaptation of the N-methyl-d-aspartate receptor complex following chronic antidepressant treatments. J Pharmacol Exp Ther. 1994;269(1):95–102.PubMed
91.
Zurück zum Zitat Nowak G, Li Y, Paul IA. Adaptation of cortical but not hippocampal NMDA receptors after chronic citalopram treatment. Eur J Pharmacol. 1996;295(1):75–85.PubMedCrossRef Nowak G, Li Y, Paul IA. Adaptation of cortical but not hippocampal NMDA receptors after chronic citalopram treatment. Eur J Pharmacol. 1996;295(1):75–85.PubMedCrossRef
92.
Zurück zum Zitat Murrough JW, Abdallah CG, Mathew SJ. Targeting glutamate signalling in depression: progress and prospects. Nat Rev Drug Discov. 2017;16(7):472–86.PubMedCrossRef Murrough JW, Abdallah CG, Mathew SJ. Targeting glutamate signalling in depression: progress and prospects. Nat Rev Drug Discov. 2017;16(7):472–86.PubMedCrossRef
93.
Zurück zum Zitat Jimenez-Sanchez L, Campa L, Auberson YP, Adell A. The role of GluN2A and GluN2B subunits on the effects of NMDA receptor antagonists in modeling schizophrenia and treating refractory depression. Neuropsychopharmacology. 2014;39(11):2673–80.PubMedPubMedCentralCrossRef Jimenez-Sanchez L, Campa L, Auberson YP, Adell A. The role of GluN2A and GluN2B subunits on the effects of NMDA receptor antagonists in modeling schizophrenia and treating refractory depression. Neuropsychopharmacology. 2014;39(11):2673–80.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Kiselycznyk C, Jury NJ, Halladay LR, Nakazawa K, Mishina M, Sprengel R, et al. NMDA receptor subunits and associated signaling molecules mediating antidepressant-related effects of NMDA-GluN2B antagonism. Behav Brain Res. 2015;287:89–95.PubMedPubMedCentralCrossRef Kiselycznyk C, Jury NJ, Halladay LR, Nakazawa K, Mishina M, Sprengel R, et al. NMDA receptor subunits and associated signaling molecules mediating antidepressant-related effects of NMDA-GluN2B antagonism. Behav Brain Res. 2015;287:89–95.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Yang Y, Cui Y, Sang K, Dong Y, Ni Z, Ma S, et al. Ketamine blocks bursting in the lateral habenula to rapidly relieve depression. Nature. 2018;554:317PubMedCrossRef Yang Y, Cui Y, Sang K, Dong Y, Ni Z, Ma S, et al. Ketamine blocks bursting in the lateral habenula to rapidly relieve depression. Nature. 2018;554:317PubMedCrossRef
97.
Zurück zum Zitat Zhu W-L, Wang S-J, Liu M-M, Shi H-S, Zhang R-X, Liu J-F, et al. Glycine site N-methyl-d-aspartate receptor antagonist 7-CTKA produces rapid antidepressant-like effects in male rats. J Psychiatry Neurosci. 2013;38(5):306–16.PubMedPubMedCentralCrossRef Zhu W-L, Wang S-J, Liu M-M, Shi H-S, Zhang R-X, Liu J-F, et al. Glycine site N-methyl-d-aspartate receptor antagonist 7-CTKA produces rapid antidepressant-like effects in male rats. J Psychiatry Neurosci. 2013;38(5):306–16.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Liu B-B, Luo L, Liu X-L, Geng D, Liu Q, Yi L-T. 7-Chlorokynurenic acid (7-CTKA) produces rapid antidepressant-like effects: through regulating hippocampal microRNA expressions involved in TrkB-ERK/Akt signaling pathways in mice exposed to chronic unpredictable mild stress. Psychopharmacology. 2015;232(3):541–50.PubMedCrossRef Liu B-B, Luo L, Liu X-L, Geng D, Liu Q, Yi L-T. 7-Chlorokynurenic acid (7-CTKA) produces rapid antidepressant-like effects: through regulating hippocampal microRNA expressions involved in TrkB-ERK/Akt signaling pathways in mice exposed to chronic unpredictable mild stress. Psychopharmacology. 2015;232(3):541–50.PubMedCrossRef
99.
Zurück zum Zitat Zhang JC, Li SX, Hashimoto K. R (−)-ketamine shows greater potency and longer lasting antidepressant effects than S (+)-ketamine. Pharmacol Biochem Behav. 2014;116:137–41.PubMedCrossRef Zhang JC, Li SX, Hashimoto K. R (−)-ketamine shows greater potency and longer lasting antidepressant effects than S (+)-ketamine. Pharmacol Biochem Behav. 2014;116:137–41.PubMedCrossRef
100.
Zurück zum Zitat Yang C, Qu Y, Fujita Y, Ren Q, Ma M, Dong C, et al. Possible role of the gut microbiota-brain axis in the antidepressant effects of (R)-ketamine in a social defeat stress model. Transl Psychiatry. 2017;7(12):1294.PubMedPubMedCentralCrossRef Yang C, Qu Y, Fujita Y, Ren Q, Ma M, Dong C, et al. Possible role of the gut microbiota-brain axis in the antidepressant effects of (R)-ketamine in a social defeat stress model. Transl Psychiatry. 2017;7(12):1294.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Sarkar A, Kabbaj M. Sex differences in effects of ketamine on behavior, spine density, and synaptic proteins in socially isolated rats. Biol Psychiatry. 2016;80(6):448–56.PubMedPubMedCentralCrossRef Sarkar A, Kabbaj M. Sex differences in effects of ketamine on behavior, spine density, and synaptic proteins in socially isolated rats. Biol Psychiatry. 2016;80(6):448–56.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Can A, Zanos P, Moaddel R, Kang HJ, Dossou KSS, Wainer IW, et al. Effects of ketamine and ketamine metabolites on evoked striatal dopamine release, dopamine receptors, and monoamine transporters. J Pharmacol Exp Ther. 2016;359(1):159–70.PubMedPubMedCentralCrossRef Can A, Zanos P, Moaddel R, Kang HJ, Dossou KSS, Wainer IW, et al. Effects of ketamine and ketamine metabolites on evoked striatal dopamine release, dopamine receptors, and monoamine transporters. J Pharmacol Exp Ther. 2016;359(1):159–70.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Zarate CA Jr, Brutsche N, Laje G, Luckenbaugh DA, Venkata SL, Ramamoorthy A, et al. Relationship of ketamine’s plasma metabolites with response, diagnosis, and side effects in major depression. Biol Psychiatry. 2012;72(4):331–8.PubMedPubMedCentralCrossRef Zarate CA Jr, Brutsche N, Laje G, Luckenbaugh DA, Venkata SL, Ramamoorthy A, et al. Relationship of ketamine’s plasma metabolites with response, diagnosis, and side effects in major depression. Biol Psychiatry. 2012;72(4):331–8.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Pham TH, Defaix C, Xu X, Deng S-X, Fabresse N, Alvarez J-C, et al. Common neurotransmission recruited in recruited in (R,S)-ketamine and (2R,6R)-hydroxynorketamine-induced sustained antidepressant-like effects. Biol Psychiatry. 2017. S0006-3223(17)32130–3. https://doi.org/10.1016/j.biopsych.2017.10.020. (epub ahead of print). Pham TH, Defaix C, Xu X, Deng S-X, Fabresse N, Alvarez J-C, et al. Common neurotransmission recruited in recruited in (R,S)-ketamine and (2R,6R)-hydroxynorketamine-induced sustained antidepressant-like effects. Biol Psychiatry. 2017. S0006-3223(17)32130–3. https://​doi.​org/​10.​1016/​j.​biopsych.​2017.​10.​020. (epub ahead of print).
105.
Zurück zum Zitat Yang C, Qu Y, Abe M, Nozawa D, Chaki S, Hashimoto K. (R)-Ketamine shows greater potency and longer lasting antidepressant effects than its metabolite (2R,6R)-hydroxynorketamine. Biol Psychiatry. 2017;82(5):e43–4.PubMedCrossRef Yang C, Qu Y, Abe M, Nozawa D, Chaki S, Hashimoto K. (R)-Ketamine shows greater potency and longer lasting antidepressant effects than its metabolite (2R,6R)-hydroxynorketamine. Biol Psychiatry. 2017;82(5):e43–4.PubMedCrossRef
106.
Zurück zum Zitat Shirayama Y, Hashimoto K. Lack of antidepressant effects of (2R,6R)-hydroxynorketamine in a rat learned helplessness model: comparison with (R)-ketamine. Int J Neuropsychopharmacol. 2018;21(1):84–8.PubMedCrossRef Shirayama Y, Hashimoto K. Lack of antidepressant effects of (2R,6R)-hydroxynorketamine in a rat learned helplessness model: comparison with (R)-ketamine. Int J Neuropsychopharmacol. 2018;21(1):84–8.PubMedCrossRef
107.
Zurück zum Zitat Zanos P, Moaddel R, Morris PJ, Georgiou P, Fischell J, Elmer GI, et al. Reply to: effects of a ketamine metabolite on synaptic NMDAR function. Nature. 2017;546(7659):E4–5.PubMedPubMedCentralCrossRef Zanos P, Moaddel R, Morris PJ, Georgiou P, Fischell J, Elmer GI, et al. Reply to: effects of a ketamine metabolite on synaptic NMDAR function. Nature. 2017;546(7659):E4–5.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Suzuki K, Nosyreva E, Hunt KW, Kavalali ET, Monteggia LM. Effects of a ketamine metabolite on synaptic NMDAR function. Nature. 2017;546(7659):E1–3.PubMedCrossRef Suzuki K, Nosyreva E, Hunt KW, Kavalali ET, Monteggia LM. Effects of a ketamine metabolite on synaptic NMDAR function. Nature. 2017;546(7659):E1–3.PubMedCrossRef
109.
Zurück zum Zitat Morris PJ, Moaddel R, Zanos P, Moore CE, Gould T, Zarate CA, et al. Synthesis and N-methyl-d-aspartate (NMDA) receptor activity of ketamine metabolites. Org Lett. 2017;19(17):4572–5.PubMedPubMedCentralCrossRef Morris PJ, Moaddel R, Zanos P, Moore CE, Gould T, Zarate CA, et al. Synthesis and N-methyl-d-aspartate (NMDA) receptor activity of ketamine metabolites. Org Lett. 2017;19(17):4572–5.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Zarate CA Jr, Singh JB, Quiroz JA, De Jesus G, Denicoff KK, Luckenbaugh DA, et al. A double-blind, placebo-controlled study of memantine in the treatment of major depression. Am J Psychiatry. 2006;163(1):153–5.PubMedCrossRef Zarate CA Jr, Singh JB, Quiroz JA, De Jesus G, Denicoff KK, Luckenbaugh DA, et al. A double-blind, placebo-controlled study of memantine in the treatment of major depression. Am J Psychiatry. 2006;163(1):153–5.PubMedCrossRef
111.
Zurück zum Zitat Lenze EJ, Skidmore ER, Begley AE, Newcomer JW, Butters MA, Whyte EM. Memantine for late-life depression and apathy after a disabling medical event: a 12-week, double-blind placebo-controlled pilot study. Int J Geriatr Psychiatry. 2012;27(9):974–80.PubMedCrossRef Lenze EJ, Skidmore ER, Begley AE, Newcomer JW, Butters MA, Whyte EM. Memantine for late-life depression and apathy after a disabling medical event: a 12-week, double-blind placebo-controlled pilot study. Int J Geriatr Psychiatry. 2012;27(9):974–80.PubMedCrossRef
112.
Zurück zum Zitat Ferguson JM, Shingleton RN. An open-label, flexible-dose study of memantine in major depressive disorder. Clin Neuropharmacol. 2007;30(3):136–44.PubMedCrossRef Ferguson JM, Shingleton RN. An open-label, flexible-dose study of memantine in major depressive disorder. Clin Neuropharmacol. 2007;30(3):136–44.PubMedCrossRef
113.
Zurück zum Zitat Moaddel R, Abdrakhmanova G, Kozak J, Jozwiak K, Toll L, Jimenez L, et al. Sub-anesthetic concentrations of (R,S)-ketamine metabolites inhibit acetylcholine-evoked currents in alpha7 nicotinic acetylcholine receptors. Eur J Pharmacol. 2013;698(1–3):228–34.PubMedCrossRef Moaddel R, Abdrakhmanova G, Kozak J, Jozwiak K, Toll L, Jimenez L, et al. Sub-anesthetic concentrations of (R,S)-ketamine metabolites inhibit acetylcholine-evoked currents in alpha7 nicotinic acetylcholine receptors. Eur J Pharmacol. 2013;698(1–3):228–34.PubMedCrossRef
114.
Zurück zum Zitat 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(9):978–85.PubMedCrossRef 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(9):978–85.PubMedCrossRef
115.
Zurück zum Zitat Sanacora G, Johnson MR, Khan A, Atkinson SD, Riesenberg RR, Schronen JP, et al. Adjunctive lanicemine (AZD6765) in patients with major depressive disorder and history of inadequate response to antidepressants: a randomized, placebo-controlled study. Neuropsychopharmacology. 2017;42(4):844–53.PubMedCrossRef Sanacora G, Johnson MR, Khan A, Atkinson SD, Riesenberg RR, Schronen JP, et al. Adjunctive lanicemine (AZD6765) in patients with major depressive disorder and history of inadequate response to antidepressants: a randomized, placebo-controlled study. Neuropsychopharmacology. 2017;42(4):844–53.PubMedCrossRef
116.
Zurück zum Zitat Preskorn SH, Baker B, Kolluri S, Menniti FS, Krams M, Landen JW. An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-d-aspartate antagonist, CP-101,606, in patients with treatment-refractory major depressive disorder. J Clin Psychopharmacol. 2008;28(6):631–7.PubMedCrossRef Preskorn SH, Baker B, Kolluri S, Menniti FS, Krams M, Landen JW. An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-d-aspartate antagonist, CP-101,606, in patients with treatment-refractory major depressive disorder. J Clin Psychopharmacol. 2008;28(6):631–7.PubMedCrossRef
117.
Zurück zum Zitat Hashimoto K. Comments on “An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-d-aspartate antagonist, CP-101,606 in patients with treatment-refractory major depressive disorder”. J Clin Psychopharmacol. 2009;29(4):411–2 (author reply 2).PubMedCrossRef Hashimoto K. Comments on “An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-d-aspartate antagonist, CP-101,606 in patients with treatment-refractory major depressive disorder”. J Clin Psychopharmacol. 2009;29(4):411–2 (author reply 2).PubMedCrossRef
118.
Zurück zum Zitat Hashimoto K, London ED. Further characterization of [3H]ifenprodil binding to sigma receptors in rat brain. Eur J Pharmacol. 1993;236(1):159–63.PubMedCrossRef Hashimoto K, London ED. Further characterization of [3H]ifenprodil binding to sigma receptors in rat brain. Eur J Pharmacol. 1993;236(1):159–63.PubMedCrossRef
119.
Zurück zum Zitat Hashimoto K, Ishiwata K. Sigma receptor ligands: possible application as therapeutic drugs and as radiopharmaceuticals. Curr Pharm Des. 2006;12(30):3857–76.PubMed Hashimoto K, Ishiwata K. Sigma receptor ligands: possible application as therapeutic drugs and as radiopharmaceuticals. Curr Pharm Des. 2006;12(30):3857–76.PubMed
120.
Zurück zum Zitat Stahl SM. The sigma enigma: can sigma receptors provide a novel target for disorders of mood and cognition? J Clin Psychiatry. 2008;69(11):1673–4.PubMedCrossRef Stahl SM. The sigma enigma: can sigma receptors provide a novel target for disorders of mood and cognition? J Clin Psychiatry. 2008;69(11):1673–4.PubMedCrossRef
121.
Zurück zum Zitat Hashimoto K. Sigma-1 receptors and selective serotonin reuptake inhibitors: clinical implications of their relationship. Cent Nerv Syst Agents Med Chem. 2009;9(3):197–204.PubMedCrossRef Hashimoto K. Sigma-1 receptors and selective serotonin reuptake inhibitors: clinical implications of their relationship. Cent Nerv Syst Agents Med Chem. 2009;9(3):197–204.PubMedCrossRef
122.
Zurück zum Zitat Ibrahim L, Diaz Granados N, Jolkovsky L, Brutsche N, Luckenbaugh DA, Herring WJ, et al. A randomized, placebo-controlled, crossover pilot trial of the oral selective NR2B antagonist MK-0657 in patients with treatment-resistant major depressive disorder. J Clin Psychopharmacol. 2012;32(4):551–7.PubMedPubMedCentralCrossRef Ibrahim L, Diaz Granados N, Jolkovsky L, Brutsche N, Luckenbaugh DA, Herring WJ, et al. A randomized, placebo-controlled, crossover pilot trial of the oral selective NR2B antagonist MK-0657 in patients with treatment-resistant major depressive disorder. J Clin Psychopharmacol. 2012;32(4):551–7.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Sanacora G. What are we learning from early-phase clinical trials with glutamate targeting medications for the treatment of major depressive disorder. JAMA Psychiatry. 2016;73(7):651–2.PubMedCrossRef Sanacora G. What are we learning from early-phase clinical trials with glutamate targeting medications for the treatment of major depressive disorder. JAMA Psychiatry. 2016;73(7):651–2.PubMedCrossRef
124.
Zurück zum Zitat Thompson SM, Kallarackal AJ, Kvarta MD, Van Dyke AM, LeGates TA, Cai X. An excitatory synapse hypothesis of depression. Trends Neurosci. 2015;38(5):279–94.PubMedPubMedCentralCrossRef Thompson SM, Kallarackal AJ, Kvarta MD, Van Dyke AM, LeGates TA, Cai X. An excitatory synapse hypothesis of depression. Trends Neurosci. 2015;38(5):279–94.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Zanos P, Gould TD. Mechanisms of ketamine action as an antidepressant. Mol Psychiatry. (in press). Zanos P, Gould TD. Mechanisms of ketamine action as an antidepressant. Mol Psychiatry. (in press).
126.
Zurück zum Zitat Moskal JR, Kuo AG, Weiss C, Wood PL, Hanson AO, Kelso S, et al. GLYX-13: a monoclonal antibody-derived peptide that acts as an N-methyl-d-aspartate receptor modulator. Neuropharmacology. 2005;49(7):1077–87.PubMedCrossRef Moskal JR, Kuo AG, Weiss C, Wood PL, Hanson AO, Kelso S, et al. GLYX-13: a monoclonal antibody-derived peptide that acts as an N-methyl-d-aspartate receptor modulator. Neuropharmacology. 2005;49(7):1077–87.PubMedCrossRef
127.
Zurück zum Zitat Preskorn S, Macaluso M, Mehra DO, Zammit G, Moskal JR, Burch RM, et al. Randomized proof of concept trial of GLYX-13, an N-methyl-d-aspartate receptor glycine site partial agonist, in major depressive disorder nonresponsive to a previous antidepressant agent. J Psychiatr Pract. 2015;21(2):140–9.PubMedCrossRef Preskorn S, Macaluso M, Mehra DO, Zammit G, Moskal JR, Burch RM, et al. Randomized proof of concept trial of GLYX-13, an N-methyl-d-aspartate receptor glycine site partial agonist, in major depressive disorder nonresponsive to a previous antidepressant agent. J Psychiatr Pract. 2015;21(2):140–9.PubMedCrossRef
128.
Zurück zum Zitat Liu RJ, Duman C, Kato T, Hare B, Lopresto D, Bang E, et al. GLYX-13 produces rapid antidepressant responses with key synaptic and behavioral effects distinct from ketamine. Neuropsychopharmacology. 2017;42(6):1231–42.PubMedCrossRef Liu RJ, Duman C, Kato T, Hare B, Lopresto D, Bang E, et al. GLYX-13 produces rapid antidepressant responses with key synaptic and behavioral effects distinct from ketamine. Neuropsychopharmacology. 2017;42(6):1231–42.PubMedCrossRef
129.
Zurück zum Zitat Chen L, Muhlhauser M, Yang CR. Glycine tranporter-1 blockade potentiates NMDA-mediated responses in rat prefrontal cortical neurons in vitro and in vivo. J Neurophysiol. 2003;89(2):691–703.PubMedCrossRef Chen L, Muhlhauser M, Yang CR. Glycine tranporter-1 blockade potentiates NMDA-mediated responses in rat prefrontal cortical neurons in vitro and in vivo. J Neurophysiol. 2003;89(2):691–703.PubMedCrossRef
130.
Zurück zum Zitat Huang CC, Wei IH, Huang CL, Chen KT, Tsai MH, Tsai P, et al. Inhibition of glycine transporter-I as a novel mechanism for the treatment of depression. Biol Psychiatry. 2013;74(10):734–41.PubMedCrossRef Huang CC, Wei IH, Huang CL, Chen KT, Tsai MH, Tsai P, et al. Inhibition of glycine transporter-I as a novel mechanism for the treatment of depression. Biol Psychiatry. 2013;74(10):734–41.PubMedCrossRef
131.
Zurück zum Zitat Heresco-Levy U, Gelfin G, Bloch B, Levin R, Edelman S, Javitt DC, et al. A randomized add-on trial of high-dose d-cycloserine for treatment-resistant depression. Int J Neuropsychopharmacol. 2013;16(3):501–6.PubMedCrossRef Heresco-Levy U, Gelfin G, Bloch B, Levin R, Edelman S, Javitt DC, et al. A randomized add-on trial of high-dose d-cycloserine for treatment-resistant depression. Int J Neuropsychopharmacol. 2013;16(3):501–6.PubMedCrossRef
132.
Zurück zum Zitat Wilhelm S, Buhlmann U, Tolin DF, Meunier SA, Pearlson GD, Reese HE, et al. Augmentation of behavior therapy with d-cycloserine for obsessive-compulsive disorder. Am J Psychiatry. 2008;165(3):335–41 (quiz 409).PubMedCrossRef Wilhelm S, Buhlmann U, Tolin DF, Meunier SA, Pearlson GD, Reese HE, et al. Augmentation of behavior therapy with d-cycloserine for obsessive-compulsive disorder. Am J Psychiatry. 2008;165(3):335–41 (quiz 409).PubMedCrossRef
133.
Zurück zum Zitat Poleszak E, Wlaź P, Szewczyk B, Wlaź A, Kasperek R, Wróbel A, et al. A complex interaction between glycine/NMDA receptors and serotonergic/noradrenergic antidepressants in the forced swim test in mice. J Neural Transm. 2011;118(11):1535–46.PubMedPubMedCentralCrossRef Poleszak E, Wlaź P, Szewczyk B, Wlaź A, Kasperek R, Wróbel A, et al. A complex interaction between glycine/NMDA receptors and serotonergic/noradrenergic antidepressants in the forced swim test in mice. J Neural Transm. 2011;118(11):1535–46.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat van Berckel BN, Lipsch C, Timp S, Gispen-de Wied C, Wynne H, van Ree JM, et al. Behavioral and neuroendocrine effects of the partial NMDA agonist d-cycloserine in healthy subjects. Neuropsychopharmacology. 1997;16(5):317–24.PubMedCrossRef van Berckel BN, Lipsch C, Timp S, Gispen-de Wied C, Wynne H, van Ree JM, et al. Behavioral and neuroendocrine effects of the partial NMDA agonist d-cycloserine in healthy subjects. Neuropsychopharmacology. 1997;16(5):317–24.PubMedCrossRef
135.
Zurück zum Zitat Trullas R, Folio T, Young A, Miller R, Boje K, Skolnick P. 1-aminocyclopropanecarboxylates exhibit antidepressant and anxiolytic actions in animal models. Eur J Pharmacol. 1991;203(3):379–85.PubMedCrossRef Trullas R, Folio T, Young A, Miller R, Boje K, Skolnick P. 1-aminocyclopropanecarboxylates exhibit antidepressant and anxiolytic actions in animal models. Eur J Pharmacol. 1991;203(3):379–85.PubMedCrossRef
136.
Zurück zum Zitat Moskal JR, Burch R, Burgdorf JS, Kroes RA, Stanton PK, Disterhoft JF, et al. GLYX-13, an NMDA receptor glycine site functional partial agonist enhances cognition and produces antidepressant effects without the psychotomimetic side effects of NMDA receptor antagonists. Expert Opin Investig Drugs. 2014;23(2):243–54.PubMedCrossRef Moskal JR, Burch R, Burgdorf JS, Kroes RA, Stanton PK, Disterhoft JF, et al. GLYX-13, an NMDA receptor glycine site functional partial agonist enhances cognition and produces antidepressant effects without the psychotomimetic side effects of NMDA receptor antagonists. Expert Opin Investig Drugs. 2014;23(2):243–54.PubMedCrossRef
137.
Zurück zum Zitat Rajagopal L, Burgdorf JS, Moskal JR, Meltzer HY. GLYX-13 (rapastinel) ameliorates subchronic phencyclidine- and ketamine-induced declarative memory deficits in mice. Behav Brain Res. 2016;15(299):105–10.CrossRef Rajagopal L, Burgdorf JS, Moskal JR, Meltzer HY. GLYX-13 (rapastinel) ameliorates subchronic phencyclidine- and ketamine-induced declarative memory deficits in mice. Behav Brain Res. 2016;15(299):105–10.CrossRef
138.
Zurück zum Zitat Shu Y, Hasenstaub A, McCormick DA. Turning on and off recurrent balanced cortical activity. Nature. 2003;423(6937):288–93.PubMedCrossRef Shu Y, Hasenstaub A, McCormick DA. Turning on and off recurrent balanced cortical activity. Nature. 2003;423(6937):288–93.PubMedCrossRef
139.
Zurück zum Zitat Markram H, Toledo-Rodriguez M, Wang Y, Gupta A, Silberberg G, Wu C. Interneurons of the neocortical inhibitory system. Nat Rev Neurosci. 2004;5(10):793–807.PubMedCrossRef Markram H, Toledo-Rodriguez M, Wang Y, Gupta A, Silberberg G, Wu C. Interneurons of the neocortical inhibitory system. Nat Rev Neurosci. 2004;5(10):793–807.PubMedCrossRef
140.
Zurück zum Zitat Williams SM, Goldman-Rakic PS, Leranth C. The synaptology of parvalbumin-immunoreactive neurons in the primate prefrontal cortex. J Comp Neurol. 1992;320(3):353–69.PubMedCrossRef Williams SM, Goldman-Rakic PS, Leranth C. The synaptology of parvalbumin-immunoreactive neurons in the primate prefrontal cortex. J Comp Neurol. 1992;320(3):353–69.PubMedCrossRef
141.
Zurück zum Zitat Cobb SR, Buhl EH, Halasy K, Paulsen O, Somogyi P. Synchronization of neuronal activity in hippocampus by individual GABAergic interneurons. Nature. 1995;378(6552):75–8.PubMedCrossRef Cobb SR, Buhl EH, Halasy K, Paulsen O, Somogyi P. Synchronization of neuronal activity in hippocampus by individual GABAergic interneurons. Nature. 1995;378(6552):75–8.PubMedCrossRef
142.
Zurück zum Zitat Miles R, Toth K, Gulyas AI, Hajos N, Freund TF. Differences between somatic and dendritic inhibition in the hippocampus. Neuron. 1996;16(4):815–23.PubMedCrossRef Miles R, Toth K, Gulyas AI, Hajos N, Freund TF. Differences between somatic and dendritic inhibition in the hippocampus. Neuron. 1996;16(4):815–23.PubMedCrossRef
143.
Zurück zum Zitat Cardin JA, Carlen M, Meletis K, Knoblich U, Zhang F, Deisseroth K, et al. Driving fast-spiking cells induces gamma rhythm and controls sensory responses. Nature. 2009;459(7247):663–7.PubMedPubMedCentralCrossRef Cardin JA, Carlen M, Meletis K, Knoblich U, Zhang F, Deisseroth K, et al. Driving fast-spiking cells induces gamma rhythm and controls sensory responses. Nature. 2009;459(7247):663–7.PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Sohal VS, Zhang F, Yizhar O, Deisseroth K. Parvalbumin neurons and gamma rhythms enhance cortical circuit performance. Nature. 2009;459(7247):698–702.PubMedPubMedCentralCrossRef Sohal VS, Zhang F, Yizhar O, Deisseroth K. Parvalbumin neurons and gamma rhythms enhance cortical circuit performance. Nature. 2009;459(7247):698–702.PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat de Lima AD, Morrison JH. Ultrastructural analysis of somatostatin-immunoreactive neurons and synapses in the temporal and occipital cortex of the macaque monkey. J Comp Neurol. 1989;283(2):212–27.PubMedCrossRef de Lima AD, Morrison JH. Ultrastructural analysis of somatostatin-immunoreactive neurons and synapses in the temporal and occipital cortex of the macaque monkey. J Comp Neurol. 1989;283(2):212–27.PubMedCrossRef
146.
Zurück zum Zitat Kawaguchi Y, Kubota Y. Physiological and morphological identification of somatostatin- or vasoactive intestinal polypeptide-containing cells among GABAergic cell subtypes in rat frontal cortex. J Neurosci. 1996;16(8):2701–15.PubMedCrossRef Kawaguchi Y, Kubota Y. Physiological and morphological identification of somatostatin- or vasoactive intestinal polypeptide-containing cells among GABAergic cell subtypes in rat frontal cortex. J Neurosci. 1996;16(8):2701–15.PubMedCrossRef
147.
Zurück zum Zitat 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(8):2921–7.PubMedCrossRef 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(8):2921–7.PubMedCrossRef
148.
Zurück zum Zitat Lorrain DS, Baccei CS, Bristow LJ, Anderson JJ, Varney MA. 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. 2003;117(3):697–706.PubMedCrossRef Lorrain DS, Baccei CS, Bristow LJ, Anderson JJ, Varney MA. 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. 2003;117(3):697–706.PubMedCrossRef
149.
Zurück zum Zitat Chowdhury GM, Zhang J, Thomas M, Banasr M, Ma X, Pittman B, et al. Transiently increased glutamate cycling in rat PFC is associated with rapid onset of antidepressant-like effects. Mol Psychiatry. 2017;22(1):120–6.PubMedCrossRef Chowdhury GM, Zhang J, Thomas M, Banasr M, Ma X, Pittman B, et al. Transiently increased glutamate cycling in rat PFC is associated with rapid onset of antidepressant-like effects. Mol Psychiatry. 2017;22(1):120–6.PubMedCrossRef
150.
Zurück zum Zitat Homayoun H, Moghaddam B. NMDA receptor hypofunction produces opposite effects on prefrontal cortex interneurons and pyramidal neurons. J Neurosci. 2007;27(43):11496–500.PubMedPubMedCentralCrossRef Homayoun H, Moghaddam B. NMDA receptor hypofunction produces opposite effects on prefrontal cortex interneurons and pyramidal neurons. J Neurosci. 2007;27(43):11496–500.PubMedPubMedCentralCrossRef
151.
Zurück zum Zitat Ling DS, Benardo LS. Recruitment of GABAA inhibition in rat neocortex is limited and not NMDA dependent. J Neurophysiol. 1995;74(6):2329–35.PubMedCrossRef Ling DS, Benardo LS. Recruitment of GABAA inhibition in rat neocortex is limited and not NMDA dependent. J Neurophysiol. 1995;74(6):2329–35.PubMedCrossRef
152.
Zurück zum Zitat Grunze HC, Rainnie DG, Hasselmo ME, Barkai E, Hearn EF, McCarley RW, et al. NMDA-dependent modulation of CA1 local circuit inhibition. J Neurosci. 1996;16(6):2034–43.PubMedCrossRef Grunze HC, Rainnie DG, Hasselmo ME, Barkai E, Hearn EF, McCarley RW, et al. NMDA-dependent modulation of CA1 local circuit inhibition. J Neurosci. 1996;16(6):2034–43.PubMedCrossRef
153.
Zurück zum Zitat Hiyoshi T, Kambe D, Karasawa J, Chaki S. Involvement of glutamatergic and GABAergic transmission in MK-801-increased gamma band oscillation power in rat cortical electroencephalograms. Neuroscience. 2014;7(280):262–74.CrossRef Hiyoshi T, Kambe D, Karasawa J, Chaki S. Involvement of glutamatergic and GABAergic transmission in MK-801-increased gamma band oscillation power in rat cortical electroencephalograms. Neuroscience. 2014;7(280):262–74.CrossRef
154.
Zurück zum Zitat 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;17(10):1671–81.PubMedCrossRef 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;17(10):1671–81.PubMedCrossRef
155.
Zurück zum Zitat Kocsis B. Differential role of NR2A and NR2B subunits in N-methyl-d-aspartate receptor antagonist-induced aberrant cortical gamma oscillations. Biol Psychiatry. 2012;71(11):987–95.PubMedCrossRef Kocsis B. Differential role of NR2A and NR2B subunits in N-methyl-d-aspartate receptor antagonist-induced aberrant cortical gamma oscillations. Biol Psychiatry. 2012;71(11):987–95.PubMedCrossRef
156.
Zurück zum Zitat Kocsis B. State-dependent increase of cortical gamma activity during REM sleep after selective blockade of NR2B subunit containing NMDA receptors. Sleep. 2012;35(7):1011–6.PubMedPubMedCentralCrossRef Kocsis B. State-dependent increase of cortical gamma activity during REM sleep after selective blockade of NR2B subunit containing NMDA receptors. Sleep. 2012;35(7):1011–6.PubMedPubMedCentralCrossRef
157.
Zurück zum Zitat Sapkota K, Mao Z, Synowicki P, Lieber D, Liu M, Ikezu T, et al. GluN2D N-methyl-d-aspartate receptor subunit contribution to the stimulation of brain activity and gamma oscillations by ketamine: implications for schizophrenia. J Pharmacol Exp Ther. 2016;356(3):702–11.PubMedPubMedCentralCrossRef Sapkota K, Mao Z, Synowicki P, Lieber D, Liu M, Ikezu T, et al. GluN2D N-methyl-d-aspartate receptor subunit contribution to the stimulation of brain activity and gamma oscillations by ketamine: implications for schizophrenia. J Pharmacol Exp Ther. 2016;356(3):702–11.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Disney AA, Reynolds JH. Expression of m1-type muscarinic acetylcholine receptors by parvalbumin-immunoreactive neurons in the primary visual cortex: a comparative study of rat, guinea pig, ferret, macaque, and human. J Comp Neurol. 2014;522(5):986–1003.PubMedPubMedCentralCrossRef Disney AA, Reynolds JH. Expression of m1-type muscarinic acetylcholine receptors by parvalbumin-immunoreactive neurons in the primary visual cortex: a comparative study of rat, guinea pig, ferret, macaque, and human. J Comp Neurol. 2014;522(5):986–1003.PubMedPubMedCentralCrossRef
159.
Zurück zum Zitat McCormick DA, Prince DA. Two types of muscarinic response to acetylcholine in mammalian cortical neurons. Proc Nat Acad Sci USA. 1985;82(18):6344–8.PubMedPubMedCentralCrossRef McCormick DA, Prince DA. Two types of muscarinic response to acetylcholine in mammalian cortical neurons. Proc Nat Acad Sci USA. 1985;82(18):6344–8.PubMedPubMedCentralCrossRef
160.
Zurück zum Zitat Amar M, Lucas-Meunier E, Baux G, Fossier P. Blockade of different muscarinic receptor subtypes changes the equilibrium between excitation and inhibition in rat visual cortex. Neuroscience. 2010;169(4):1610–20.PubMedCrossRef Amar M, Lucas-Meunier E, Baux G, Fossier P. Blockade of different muscarinic receptor subtypes changes the equilibrium between excitation and inhibition in rat visual cortex. Neuroscience. 2010;169(4):1610–20.PubMedCrossRef
161.
Zurück zum Zitat Towers SK, Gloveli T, Traub RD, Driver JE, Engel D, Fradley R, et al. Alpha 5 subunit-containing GABAA receptors affect the dynamic range of mouse hippocampal kainate-induced gamma frequency oscillations in vitro. J Physiol. 2004;559(Pt 3):721–8.PubMedPubMedCentralCrossRef Towers SK, Gloveli T, Traub RD, Driver JE, Engel D, Fradley R, et al. Alpha 5 subunit-containing GABAA receptors affect the dynamic range of mouse hippocampal kainate-induced gamma frequency oscillations in vitro. J Physiol. 2004;559(Pt 3):721–8.PubMedPubMedCentralCrossRef
162.
Zurück zum Zitat Fischell J, Van Dyke AM, Kvarta MD, LeGates TA, Thompson SM. Rapid antidepressant action and restoration of excitatory synaptic strength after chronic stress by negative modulators of alpha5-containing GABAA receptors. Neuropsychopharmacology. 2015;40(11):2499–509.PubMedPubMedCentralCrossRef Fischell J, Van Dyke AM, Kvarta MD, LeGates TA, Thompson SM. Rapid antidepressant action and restoration of excitatory synaptic strength after chronic stress by negative modulators of alpha5-containing GABAA receptors. Neuropsychopharmacology. 2015;40(11):2499–509.PubMedPubMedCentralCrossRef
164.
Zurück zum Zitat Zanos P, Nelson ME, Highland JN, Krimmel SR, Georgiou P, Gould TD, et al. A negative allosteric modulator for alpha5 subunit-containing GABA receptors exerts a rapid and persistent antidepressant-like action without the side effects of the NMDA receptor antagonist ketamine in mice. eNeuro. 2017;4(1). https://doi.org/10.1523/eneuro.0285-16.2017. Zanos P, Nelson ME, Highland JN, Krimmel SR, Georgiou P, Gould TD, et al. A negative allosteric modulator for alpha5 subunit-containing GABA receptors exerts a rapid and persistent antidepressant-like action without the side effects of the NMDA receptor antagonist ketamine in mice. eNeuro. 2017;4(1). https://​doi.​org/​10.​1523/​eneuro.​0285-16.​2017.
165.
Zurück zum Zitat Xu NZ, Ernst M, Treven M, Cerne R, Wakulchik M, Li X, et al. Negative allosteric modulation of alpha 5-containing GABAA receptors engenders antidepressant-like effects and selectively prevents age-associated hyperactivity in tau-depositing mice. Psychopharmacology. 2018. https://doi.org/10.1007/s00213-018-4832-9 Xu NZ, Ernst M, Treven M, Cerne R, Wakulchik M, Li X, et al. Negative allosteric modulation of alpha 5-containing GABAA receptors engenders antidepressant-like effects and selectively prevents age-associated hyperactivity in tau-depositing mice. Psychopharmacology. 2018. https://​doi.​org/​10.​1007/​s00213-018-4832-9
166.
Zurück zum Zitat Atack JR, Maubach KA, Wafford KA, O’Connor D, Rodrigues AD, Evans DC, et al. In vitro and in vivo properties of 3-tert-butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)- pyrazolo[1,5-d]-[1,2,4]triazine (MRK-016), a GABAA receptor alpha5 subtype-selective inverse agonist. J Pharmacol Exp Ther. 2009;331(2):470–84.PubMedCrossRef Atack JR, Maubach KA, Wafford KA, O’Connor D, Rodrigues AD, Evans DC, et al. In vitro and in vivo properties of 3-tert-butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)- pyrazolo[1,5-d]-[1,2,4]triazine (MRK-016), a GABAA receptor alpha5 subtype-selective inverse agonist. J Pharmacol Exp Ther. 2009;331(2):470–84.PubMedCrossRef
167.
Zurück zum Zitat Malherbe P, Sigel E, Baur R, Persohn E, Richards JG, Mohler H. Functional expression and sites of gene transcription of a novel alpha subunit of the GABAA receptor in rat brain. FEBS Lett. 1990;260(2):261–5.PubMedCrossRef Malherbe P, Sigel E, Baur R, Persohn E, Richards JG, Mohler H. Functional expression and sites of gene transcription of a novel alpha subunit of the GABAA receptor in rat brain. FEBS Lett. 1990;260(2):261–5.PubMedCrossRef
168.
Zurück zum Zitat Lingford-Hughes A, Hume SP, Feeney A, Hirani E, Osman S, Cunningham VJ, et al. Imaging the GABA-benzodiazepine receptor subtype containing the alpha5-subunit in vivo with [11C]Ro15 4513 positron emission tomography. J Cereb Blood Flow Metab. 2002;22(7):878–89.PubMedCrossRef Lingford-Hughes A, Hume SP, Feeney A, Hirani E, Osman S, Cunningham VJ, et al. Imaging the GABA-benzodiazepine receptor subtype containing the alpha5-subunit in vivo with [11C]Ro15 4513 positron emission tomography. J Cereb Blood Flow Metab. 2002;22(7):878–89.PubMedCrossRef
169.
Zurück zum Zitat 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;80(6):457–68.PubMedPubMedCentralCrossRef 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;80(6):457–68.PubMedPubMedCentralCrossRef
170.
Zurück zum Zitat Shen Q, Lal R, Luellen BA, Earnheart JC, Andrews AM, Luscher B. γ-Aminobutyric acid-type A receptor deficits cause hypothalamic-pituitary-adrenal axis hyperactivity and antidepressant drug sensitivity reminiscent of melancholic forms of depression. Biol Psychiatry. 2010;68(6):512–20.PubMedPubMedCentralCrossRef Shen Q, Lal R, Luellen BA, Earnheart JC, Andrews AM, Luscher B. γ-Aminobutyric acid-type A receptor deficits cause hypothalamic-pituitary-adrenal axis hyperactivity and antidepressant drug sensitivity reminiscent of melancholic forms of depression. Biol Psychiatry. 2010;68(6):512–20.PubMedPubMedCentralCrossRef
171.
Zurück zum Zitat Smith KS, Rudolph U. Anxiety and depression: mouse genetics and pharmacological approaches to the role of GABA(A) receptor subtypes. Neuropharmacology. 2012;62(1):54–62.PubMedCrossRef Smith KS, Rudolph U. Anxiety and depression: mouse genetics and pharmacological approaches to the role of GABA(A) receptor subtypes. Neuropharmacology. 2012;62(1):54–62.PubMedCrossRef
172.
Zurück zum Zitat Fuchs T, Jefferson SJ, Hooper A, Yee P-HP, Maguire J, Luscher B. Disinhibition of somatostatin-positive GABAergic interneurons results in an anxiolytic and antidepressant-like brain state. Mol Psychiatry. 2017;22(6):920–30.PubMedCrossRef Fuchs T, Jefferson SJ, Hooper A, Yee P-HP, Maguire J, Luscher B. Disinhibition of somatostatin-positive GABAergic interneurons results in an anxiolytic and antidepressant-like brain state. Mol Psychiatry. 2017;22(6):920–30.PubMedCrossRef
173.
Zurück zum Zitat Möhler H. The GABA system in anxiety and depression and its therapeutic potential. Neuropharmacology. 2012;62(1):42–53.PubMedCrossRef Möhler H. The GABA system in anxiety and depression and its therapeutic potential. Neuropharmacology. 2012;62(1):42–53.PubMedCrossRef
174.
Zurück zum Zitat Kalueff AV, Nutt DJ. Role of GABA in anxiety and depression. Depress Anxiety. 2007;24(7):495–517.PubMedCrossRef Kalueff AV, Nutt DJ. Role of GABA in anxiety and depression. Depress Anxiety. 2007;24(7):495–517.PubMedCrossRef
175.
Zurück zum Zitat Klumpers UM, Veltman DJ, Drent ML, Boellaard R, Comans EF, Meynen G, et al. Reduced parahippocampal and lateral temporal GABAA-[11C]flumazenil binding in major depression: preliminary results. Eur J Nucl Med Mol Imaging. 2010;37(3):565–74.PubMedCrossRef Klumpers UM, Veltman DJ, Drent ML, Boellaard R, Comans EF, Meynen G, et al. Reduced parahippocampal and lateral temporal GABAA-[11C]flumazenil binding in major depression: preliminary results. Eur J Nucl Med Mol Imaging. 2010;37(3):565–74.PubMedCrossRef
176.
Zurück zum Zitat Sanacora G, Mason GF, Rothman DL, Krystal JH. Increased occipital cortex GABA concentrations in depressed patients after therapy with selective serotonin reuptake inhibitors. Am J Psychiatry. 2002;159(4):663–5.PubMedCrossRef Sanacora G, Mason GF, Rothman DL, Krystal JH. Increased occipital cortex GABA concentrations in depressed patients after therapy with selective serotonin reuptake inhibitors. Am J Psychiatry. 2002;159(4):663–5.PubMedCrossRef
177.
Zurück zum Zitat Fava M, Schaefer K, Huang H, Wilson A, Iosifescu DV, Mischoulon D, et al. A post hoc analysis of the effect of nightly administration of eszopiclone and a selective serotonin reuptake inhibitor in patients with insomnia and anxious depression. J Clin Psychiatry. 2011;72(4):473–9.PubMedCrossRef Fava M, Schaefer K, Huang H, Wilson A, Iosifescu DV, Mischoulon D, et al. A post hoc analysis of the effect of nightly administration of eszopiclone and a selective serotonin reuptake inhibitor in patients with insomnia and anxious depression. J Clin Psychiatry. 2011;72(4):473–9.PubMedCrossRef
178.
Zurück zum Zitat Distler MG, Plant LD, Sokoloff G, Hawk AJ, Aneas I, Wuenschell GE, et al. Glyoxalase 1 increases anxiety by reducing GABAA receptor agonist methylglyoxal. J Clin Invest. 2012;122(6):2306–15.PubMedPubMedCentralCrossRef Distler MG, Plant LD, Sokoloff G, Hawk AJ, Aneas I, Wuenschell GE, et al. Glyoxalase 1 increases anxiety by reducing GABAA receptor agonist methylglyoxal. J Clin Invest. 2012;122(6):2306–15.PubMedPubMedCentralCrossRef
180.
Zurück zum Zitat Piantadosi SC, French BJ, Poe MM, Timic T, Markovic BD, Pabba M, et al. Sex-dependent anti-stress effect of an alpha5 subunit containing GABAA receptor positive allosteric modulator. Front Pharmacol. 2016;7:446.PubMedPubMedCentralCrossRef Piantadosi SC, French BJ, Poe MM, Timic T, Markovic BD, Pabba M, et al. Sex-dependent anti-stress effect of an alpha5 subunit containing GABAA receptor positive allosteric modulator. Front Pharmacol. 2016;7:446.PubMedPubMedCentralCrossRef
181.
Zurück zum Zitat Bespalov AY, van Gaalen MM, Sukhotina IA, Wicke K, Mezler M, Schoemaker H, et al. Behavioral characterization of the mGlu group II/III receptor antagonist, LY-341495, in animal models of anxiety and depression. Eur J Pharmacol. 2008;592(1):96–102.PubMedCrossRef Bespalov AY, van Gaalen MM, Sukhotina IA, Wicke K, Mezler M, Schoemaker H, et al. Behavioral characterization of the mGlu group II/III receptor antagonist, LY-341495, in animal models of anxiety and depression. Eur J Pharmacol. 2008;592(1):96–102.PubMedCrossRef
182.
Zurück zum Zitat Chaki S, Yoshikawa R, Hirota S, Shimazaki T, Maeda M, Kawashima N, et al. MGS0039: a potent and selective group II metabotropic glutamate receptor antagonist with antidepressant-like activity. Neuropharmacology. 2004;46(4):457–67.PubMedCrossRef Chaki S, Yoshikawa R, Hirota S, Shimazaki T, Maeda M, Kawashima N, et al. MGS0039: a potent and selective group II metabotropic glutamate receptor antagonist with antidepressant-like activity. Neuropharmacology. 2004;46(4):457–67.PubMedCrossRef
183.
Zurück zum Zitat Witkin JM, Monn JA, Schoepp DD, Li X, Overshiner C, Mitchell SN, et al. The rapidly acting antidepressant ketamine and the mGlu2/3 receptor antagonist LY341495 rapidly engage dopaminergic mood circuits. J Pharmacol Exp Ther. 2016;358(1):71–82.PubMedCrossRef Witkin JM, Monn JA, Schoepp DD, Li X, Overshiner C, Mitchell SN, et al. The rapidly acting antidepressant ketamine and the mGlu2/3 receptor antagonist LY341495 rapidly engage dopaminergic mood circuits. J Pharmacol Exp Ther. 2016;358(1):71–82.PubMedCrossRef
184.
Zurück zum Zitat Yoshimizu T, Shimazaki T, Ito A, Chaki S. An mGluR2/3 antagonist, MGS0039, exerts antidepressant and anxiolytic effects in behavioral models in rats. Psychopharmacology. 2006;186(4):587.PubMedCrossRef Yoshimizu T, Shimazaki T, Ito A, Chaki S. An mGluR2/3 antagonist, MGS0039, exerts antidepressant and anxiolytic effects in behavioral models in rats. Psychopharmacology. 2006;186(4):587.PubMedCrossRef
185.
Zurück zum Zitat Ohishi H, Ogawa-Meguro R, Shigemoto R, Kaneko T, Nakanishi S, Mizuno N. Immunohistochemical localization of metabotropic glutamate receptors, mGluR2 and mGluR3, in rat cerebellar cortex. Neuron. 1994;13(1):55–66.PubMedCrossRef Ohishi H, Ogawa-Meguro R, Shigemoto R, Kaneko T, Nakanishi S, Mizuno N. Immunohistochemical localization of metabotropic glutamate receptors, mGluR2 and mGluR3, in rat cerebellar cortex. Neuron. 1994;13(1):55–66.PubMedCrossRef
186.
Zurück zum Zitat Petralia RS, Wang YX, Niedzielski AS, Wenthold RJ. The metabotropic glutamate receptors, MGLUR2 and MGLUR3, show unique postsynaptic, presynaptic and glial localizations. Neuroscience. 1996;71(4):949–76.PubMedCrossRef Petralia RS, Wang YX, Niedzielski AS, Wenthold RJ. The metabotropic glutamate receptors, MGLUR2 and MGLUR3, show unique postsynaptic, presynaptic and glial localizations. Neuroscience. 1996;71(4):949–76.PubMedCrossRef
187.
Zurück zum Zitat Shigemoto R, Kinoshita A, Wada E, Nomura S, Ohishi H, Takada M, et al. Differential presynaptic localization of metabotropic glutamate receptor subtypes in the rat hippocampus. J Neurosci. 1997;17(19):7503–22.PubMedCrossRef Shigemoto R, Kinoshita A, Wada E, Nomura S, Ohishi H, Takada M, et al. Differential presynaptic localization of metabotropic glutamate receptor subtypes in the rat hippocampus. J Neurosci. 1997;17(19):7503–22.PubMedCrossRef
188.
Zurück zum Zitat Chen Y-L, Huang C-C, Hsu K-S. Time-dependent reversal of long-term potentiation by low-frequency stimulation at the hippocampal mossy fiber–CA3 synapses. J Neurosci. 2001;21(11):3705–14.PubMedCrossRef Chen Y-L, Huang C-C, Hsu K-S. Time-dependent reversal of long-term potentiation by low-frequency stimulation at the hippocampal mossy fiber–CA3 synapses. J Neurosci. 2001;21(11):3705–14.PubMedCrossRef
189.
Zurück zum Zitat Tzounopoulos T, Janz R, Südhof TC, Nicoll RA, Malenka RC. A role for cAMP in long-term depression at hippocampal mossy fiber synapses. Neuron. 1998;21(4):837–45.PubMedCrossRef Tzounopoulos T, Janz R, Südhof TC, Nicoll RA, Malenka RC. A role for cAMP in long-term depression at hippocampal mossy fiber synapses. Neuron. 1998;21(4):837–45.PubMedCrossRef
190.
Zurück zum Zitat Ohishi H, Shigemoto R, Nakanishi S, Mizuno N. Distribution of the mRNA for a metabotropic glutamate receptor (mGluR3) in the rat brain: an in situ hybridization study. J Comp Neurol. 1993;335(2):252–66.PubMedCrossRef Ohishi H, Shigemoto R, Nakanishi S, Mizuno N. Distribution of the mRNA for a metabotropic glutamate receptor (mGluR3) in the rat brain: an in situ hybridization study. J Comp Neurol. 1993;335(2):252–66.PubMedCrossRef
191.
Zurück zum Zitat Ciccarelli R, Sureda FX, Casabona G, Di Iorio P, Caruso A, Spinella F, et al. Opposite influence of the metabotropic glutamate receptor subtypes mGlu3 and -5 on astrocyte proliferation in culture. Glia. 1997;21(4):390–8.PubMedCrossRef Ciccarelli R, Sureda FX, Casabona G, Di Iorio P, Caruso A, Spinella F, et al. Opposite influence of the metabotropic glutamate receptor subtypes mGlu3 and -5 on astrocyte proliferation in culture. Glia. 1997;21(4):390–8.PubMedCrossRef
192.
Zurück zum Zitat Aronica E, Gorter JA, Ijlst-Keizers H, Rozemuller AJ, Yankaya B, Leenstra S, et al. Expression and functional role of mGluR3 and mGluR5 in human astrocytes and glioma cells: opposite regulation of glutamate transporter proteins. Eur J Neurosci. 2003;17(10):2106–18.PubMedCrossRef Aronica E, Gorter JA, Ijlst-Keizers H, Rozemuller AJ, Yankaya B, Leenstra S, et al. Expression and functional role of mGluR3 and mGluR5 in human astrocytes and glioma cells: opposite regulation of glutamate transporter proteins. Eur J Neurosci. 2003;17(10):2106–18.PubMedCrossRef
193.
Zurück zum Zitat Dwyer JM, Lepack AE, Duman RS. mTOR activation is required for the antidepressant effects of mGluR(2)/(3) blockade. Int J Neuropsychopharmacol. 2012;15(4):429–34.PubMedCrossRef Dwyer JM, Lepack AE, Duman RS. mTOR activation is required for the antidepressant effects of mGluR(2)/(3) blockade. Int J Neuropsychopharmacol. 2012;15(4):429–34.PubMedCrossRef
194.
Zurück zum Zitat Koike H, Fukumoto K, Iijima M, Chaki S. Role of BDNF/TrkB signaling in antidepressant-like effects of a group II metabotropic glutamate receptor antagonist in animal models of depression. Behav Brain Res. 2013;1(238):48–52.CrossRef Koike H, Fukumoto K, Iijima M, Chaki S. Role of BDNF/TrkB signaling in antidepressant-like effects of a group II metabotropic glutamate receptor antagonist in animal models of depression. Behav Brain Res. 2013;1(238):48–52.CrossRef
195.
Zurück zum Zitat Fukumoto K, Iijima M, Chaki S. Serotonin-1A receptor stimulation mediates effects of a metabotropic glutamate 2/3 receptor antagonist, 2S-2-amino-2-(1S,2S-2-carboxycycloprop-1-yl)-3-(xanth-9-yl)propanoic acid (LY341495), and an N-methyl-d-aspartate receptor antagonist, ketamine, in the novelty-suppressed feeding test. Psychopharmacology (Berl). 2014;231(11):2291–8.PubMedCrossRef Fukumoto K, Iijima M, Chaki S. Serotonin-1A receptor stimulation mediates effects of a metabotropic glutamate 2/3 receptor antagonist, 2S-2-amino-2-(1S,2S-2-carboxycycloprop-1-yl)-3-(xanth-9-yl)propanoic acid (LY341495), and an N-methyl-d-aspartate receptor antagonist, ketamine, in the novelty-suppressed feeding test. Psychopharmacology (Berl). 2014;231(11):2291–8.PubMedCrossRef
196.
Zurück zum Zitat Dwyer JM, Lepack AE, Duman RS. mGluR2/3 blockade produces rapid and long-lasting reversal of anhedonia caused by chronic stress exposure. J Mol Psychiatry. 2013;1(1):15.PubMedPubMedCentralCrossRef Dwyer JM, Lepack AE, Duman RS. mGluR2/3 blockade produces rapid and long-lasting reversal of anhedonia caused by chronic stress exposure. J Mol Psychiatry. 2013;1(1):15.PubMedPubMedCentralCrossRef
197.
Zurück zum Zitat Ago Y, Yano K, Araki R, Hiramatsu N, Kita Y, Kawasaki T, et al. Metabotropic glutamate 2/3 receptor antagonists improve behavioral and prefrontal dopaminergic alterations in the chronic corticosterone-induced depression model in mice. Neuropharmacology. 2013;65:29–38.PubMedCrossRef Ago Y, Yano K, Araki R, Hiramatsu N, Kita Y, Kawasaki T, et al. Metabotropic glutamate 2/3 receptor antagonists improve behavioral and prefrontal dopaminergic alterations in the chronic corticosterone-induced depression model in mice. Neuropharmacology. 2013;65:29–38.PubMedCrossRef
198.
Zurück zum Zitat Dong C, J-c Zhang, Yao W, Ren Q, Ma M, Yang C, et al. Rapid and sustained antidepressant action of the mGlu2/3 receptor antagonist MGS0039 in the social defeat stress model: comparison with ketamine. Int J Neuropsychopharmacol. 2017;20(3):228–36.PubMed Dong C, J-c Zhang, Yao W, Ren Q, Ma M, Yang C, et al. Rapid and sustained antidepressant action of the mGlu2/3 receptor antagonist MGS0039 in the social defeat stress model: comparison with ketamine. Int J Neuropsychopharmacol. 2017;20(3):228–36.PubMed
199.
Zurück zum Zitat Umbricht D, Niggli M, Sanwald-Ducray P, Deptula D, Moore R, Grünbauer W, et al. Results of a double-blind placebo-controlled study of the antidepressant effects of the mGLU2 negative allosteric modulator RG1578. Eur Neuropsychopharmacol. 2015;25:S447.CrossRef Umbricht D, Niggli M, Sanwald-Ducray P, Deptula D, Moore R, Grünbauer W, et al. Results of a double-blind placebo-controlled study of the antidepressant effects of the mGLU2 negative allosteric modulator RG1578. Eur Neuropsychopharmacol. 2015;25:S447.CrossRef
200.
Zurück zum Zitat Dudek SM, Bear MF. Homosynaptic long-term depression in area CA1 of hippocampus and effects of N-methyl-d-aspartate receptor blockade. Proc Natl Acad Sci USA. 1992;89(10):4363–7.PubMedPubMedCentralCrossRef Dudek SM, Bear MF. Homosynaptic long-term depression in area CA1 of hippocampus and effects of N-methyl-d-aspartate receptor blockade. Proc Natl Acad Sci USA. 1992;89(10):4363–7.PubMedPubMedCentralCrossRef
201.
Zurück zum Zitat Bliss TV, Lomo T. Long-lasting potentiation of synaptic transmission in the dentate area of the anaesthetized rabbit following stimulation of the perforant path. J Physiol. 1973;232(2):331–56.PubMedPubMedCentralCrossRef Bliss TV, Lomo T. Long-lasting potentiation of synaptic transmission in the dentate area of the anaesthetized rabbit following stimulation of the perforant path. J Physiol. 1973;232(2):331–56.PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Nicoll RA, Malenka RC. Contrasting properties of two forms of long-term potentiation in the hippocampus. Nature. 1995;377(6545):115–8.PubMedCrossRef Nicoll RA, Malenka RC. Contrasting properties of two forms of long-term potentiation in the hippocampus. Nature. 1995;377(6545):115–8.PubMedCrossRef
203.
Zurück zum Zitat Alfarez DN, Joels M, Krugers HJ. Chronic unpredictable stress impairs long-term potentiation in rat hippocampal CA1 area and dentate gyrus in vitro. Eur J Neurosci. 2003;17(9):1928–34.PubMedCrossRef Alfarez DN, Joels M, Krugers HJ. Chronic unpredictable stress impairs long-term potentiation in rat hippocampal CA1 area and dentate gyrus in vitro. Eur J Neurosci. 2003;17(9):1928–34.PubMedCrossRef
204.
Zurück zum Zitat Joels M, Karst H, Alfarez D, Heine VM, Qin Y, van Riel E, et al. Effects of chronic stress on structure and cell function in rat hippocampus and hypothalamus. Stress. 2004;7(4):221–31.PubMedCrossRef Joels M, Karst H, Alfarez D, Heine VM, Qin Y, van Riel E, et al. Effects of chronic stress on structure and cell function in rat hippocampus and hypothalamus. Stress. 2004;7(4):221–31.PubMedCrossRef
205.
Zurück zum Zitat Pavlides C, Nivon LG, McEwen BS. Effects of chronic stress on hippocampal long-term potentiation. Hippocampus. 2002;12(2):245–57.PubMedCrossRef Pavlides C, Nivon LG, McEwen BS. Effects of chronic stress on hippocampal long-term potentiation. Hippocampus. 2002;12(2):245–57.PubMedCrossRef
206.
Zurück zum Zitat Duman RS, Aghajanian GK, Sanacora G, Krystal JH. Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants. Nat Med. 2016;22(3):238–49.PubMedPubMedCentralCrossRef Duman RS, Aghajanian GK, Sanacora G, Krystal JH. Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants. Nat Med. 2016;22(3):238–49.PubMedPubMedCentralCrossRef
207.
Zurück zum Zitat Wang M, Yang Y, Dong Z, Cao J, Xu L. NR2B-containing N-methyl-d-aspartate subtype glutamate receptors regulate the acute stress effect on hippocampal long-term potentiation/long-term depression in vivo. Neuroreport. 2006;17(12):1343–6.PubMedCrossRef Wang M, Yang Y, Dong Z, Cao J, Xu L. NR2B-containing N-methyl-d-aspartate subtype glutamate receptors regulate the acute stress effect on hippocampal long-term potentiation/long-term depression in vivo. Neuroreport. 2006;17(12):1343–6.PubMedCrossRef
208.
Zurück zum Zitat Leuner B, Shors TJ. Stress, anxiety, and dendritic spines: what are the connections? Neuroscience. 2013;22(251):108–19.CrossRef Leuner B, Shors TJ. Stress, anxiety, and dendritic spines: what are the connections? Neuroscience. 2013;22(251):108–19.CrossRef
209.
Zurück zum Zitat Kallarackal AJ, Kvarta MD, Cammarata E, Jaberi L, Cai X, Bailey AM, et al. Chronic stress induces a selective decrease in AMPA receptor-mediated synaptic excitation at hippocampal temporoammonic-CA1 synapses. J Neurosci. 2013;33(40):15669–74.PubMedPubMedCentralCrossRef Kallarackal AJ, Kvarta MD, Cammarata E, Jaberi L, Cai X, Bailey AM, et al. Chronic stress induces a selective decrease in AMPA receptor-mediated synaptic excitation at hippocampal temporoammonic-CA1 synapses. J Neurosci. 2013;33(40):15669–74.PubMedPubMedCentralCrossRef
211.
Zurück zum Zitat Cai X, Kallarackal AJ, Kvarta MD, Goluskin S, Gaylor K, Bailey AM, et al. Local potentiation of excitatory synapses by serotonin and its alteration in rodent models of depression. Nat Neurosci. 2013;16(4):464–72.PubMedPubMedCentralCrossRef Cai X, Kallarackal AJ, Kvarta MD, Goluskin S, Gaylor K, Bailey AM, et al. Local potentiation of excitatory synapses by serotonin and its alteration in rodent models of depression. Nat Neurosci. 2013;16(4):464–72.PubMedPubMedCentralCrossRef
212.
213.
Zurück zum Zitat Yao N, Skiteva O, Zhang X, Svenningsson P, Chergui K. Ketamine and its metabolite (2R,6R)-hydroxynorketamine induce lasting alterations in glutamatergic synaptic plasticity in the mesolimbic circuit. Mol Psychiatry. 2017. https://doi.org/10.1038/mp.2017.239. (epub ahead of print). Yao N, Skiteva O, Zhang X, Svenningsson P, Chergui K. Ketamine and its metabolite (2R,6R)-hydroxynorketamine induce lasting alterations in glutamatergic synaptic plasticity in the mesolimbic circuit. Mol Psychiatry. 2017. https://​doi.​org/​10.​1038/​mp.​2017.​239. (epub ahead of print).
214.
Zurück zum Zitat Moaddel R, Sanghvi M, Dossou KS, Ramamoorthy A, Green C, Bupp J, et al. The distribution and clearance of (2S,6S)-hydroxynorketamine, an active ketamine metabolite, in Wistar rats. Pharmacol Res Perspect. 2015;3(4):e00157.PubMedPubMedCentralCrossRef Moaddel R, Sanghvi M, Dossou KS, Ramamoorthy A, Green C, Bupp J, et al. The distribution and clearance of (2S,6S)-hydroxynorketamine, an active ketamine metabolite, in Wistar rats. Pharmacol Res Perspect. 2015;3(4):e00157.PubMedPubMedCentralCrossRef
215.
Zurück zum Zitat Zhang XL, Sullivan JA, Moskal JR, Stanton PK. A NMDA receptor glycine site partial agonist, GLYX-13, simultaneously enhances LTP and reduces LTD at Schaffer collateral-CA1 synapses in hippocampus. Neuropharmacology. 2008;55(7):1238–50.PubMedPubMedCentralCrossRef Zhang XL, Sullivan JA, Moskal JR, Stanton PK. A NMDA receptor glycine site partial agonist, GLYX-13, simultaneously enhances LTP and reduces LTD at Schaffer collateral-CA1 synapses in hippocampus. Neuropharmacology. 2008;55(7):1238–50.PubMedPubMedCentralCrossRef
216.
Zurück zum Zitat Burgdorf J, Zhang XL, Weiss C, Matthews E, Disterhoft JF, Stanton PK, et al. The N-methyl-d-aspartate receptor modulator GLYX-13 enhances learning and memory, in young adult and learning impaired aging rats. Neurobiol Aging. 2011;32(4):698–706.PubMedCrossRef Burgdorf J, Zhang XL, Weiss C, Matthews E, Disterhoft JF, Stanton PK, et al. The N-methyl-d-aspartate receptor modulator GLYX-13 enhances learning and memory, in young adult and learning impaired aging rats. Neurobiol Aging. 2011;32(4):698–706.PubMedCrossRef
217.
Zurück zum Zitat Burgdorf J, Kroes RA, X-l Zhang, Gross AL, Schmidt M, Weiss C, et al. Rapastinel (GLYX-13) has therapeutic potential for the treatment of post-traumatic stress disorder: characterization of a NMDA receptor-mediated metaplasticity process in the medial prefrontal cortex of rats. Behav Brain Res. 2015;294:177–85.PubMedPubMedCentralCrossRef Burgdorf J, Kroes RA, X-l Zhang, Gross AL, Schmidt M, Weiss C, et al. Rapastinel (GLYX-13) has therapeutic potential for the treatment of post-traumatic stress disorder: characterization of a NMDA receptor-mediated metaplasticity process in the medial prefrontal cortex of rats. Behav Brain Res. 2015;294:177–85.PubMedPubMedCentralCrossRef
218.
Zurück zum Zitat Burgdorf J, X-l Zhang, Weiss C, Gross A, Boikess SR, Kroes RA, et al. The long-lasting antidepressant effects of rapastinel (GLYX-13) are associated with a metaplasticity process in the medial prefrontal cortex and hippocampus. Neuroscience. 2015;308:202–11.PubMedPubMedCentralCrossRef Burgdorf J, X-l Zhang, Weiss C, Gross A, Boikess SR, Kroes RA, et al. The long-lasting antidepressant effects of rapastinel (GLYX-13) are associated with a metaplasticity process in the medial prefrontal cortex and hippocampus. Neuroscience. 2015;308:202–11.PubMedPubMedCentralCrossRef
219.
Zurück zum Zitat Pöschel B, Wroblewska B, Heinemann U, Manahan-Vaughan D. The metabotropic glutamate receptor mGluR3 is critically required for hippocampal long-term depression and modulates long-term potentiation in the dentate gyrus of freely moving rats. Cereb Cortex. 2005;15(9):1414–23.PubMedCrossRef Pöschel B, Wroblewska B, Heinemann U, Manahan-Vaughan D. The metabotropic glutamate receptor mGluR3 is critically required for hippocampal long-term depression and modulates long-term potentiation in the dentate gyrus of freely moving rats. Cereb Cortex. 2005;15(9):1414–23.PubMedCrossRef
220.
Zurück zum Zitat Hascup ER, Hascup KN, Stephens M, Pomerleau F, Huettl P, Gratton A, et al. Rapid microelectrode measurements and the origin and regulation of extracellular glutamate in rat prefrontal cortex. J Neurochem. 2010;115(6):1608–20.PubMedPubMedCentralCrossRef Hascup ER, Hascup KN, Stephens M, Pomerleau F, Huettl P, Gratton A, et al. Rapid microelectrode measurements and the origin and regulation of extracellular glutamate in rat prefrontal cortex. J Neurochem. 2010;115(6):1608–20.PubMedPubMedCentralCrossRef
221.
Zurück zum Zitat Chana G, Landau S, Beasley C, Everall IP, Cotter D. Two-dimensional assessment of cytoarchitecture in the anterior cingulate cortex in major depressive disorder, bipolar disorder, and schizophrenia: evidence for decreased neuronal somal size and increased neuronal density. Biol Psychiatry. 2003;53(12):1086–98.PubMedCrossRef Chana G, Landau S, Beasley C, Everall IP, Cotter D. Two-dimensional assessment of cytoarchitecture in the anterior cingulate cortex in major depressive disorder, bipolar disorder, and schizophrenia: evidence for decreased neuronal somal size and increased neuronal density. Biol Psychiatry. 2003;53(12):1086–98.PubMedCrossRef
222.
Zurück zum Zitat Cotter D, Mackay D, Chana G, Beasley C, Landau S, Everall IP. Reduced neuronal size and glial cell density in area 9 of the dorsolateral prefrontal cortex in subjects with major depressive disorder. Cereb Cortex. 2002;12(4):386–94.PubMedCrossRef Cotter D, Mackay D, Chana G, Beasley C, Landau S, Everall IP. Reduced neuronal size and glial cell density in area 9 of the dorsolateral prefrontal cortex in subjects with major depressive disorder. Cereb Cortex. 2002;12(4):386–94.PubMedCrossRef
223.
Zurück zum Zitat Cotter D, Mackay D, Landau S, Kerwin R, Everall I. Reduced glial cell density and neuronal size in the anterior cingulate cortex in major depressive disorder. Arch Gen Psychiatry. 2001;58(6):545–53.PubMedCrossRef Cotter D, Mackay D, Landau S, Kerwin R, Everall I. Reduced glial cell density and neuronal size in the anterior cingulate cortex in major depressive disorder. Arch Gen Psychiatry. 2001;58(6):545–53.PubMedCrossRef
224.
Zurück zum Zitat Cotter D, Hudson L, Landau S. Evidence for orbitofrontal pathology in bipolar disorder and major depression, but not in schizophrenia. Bipolar Disord. 2005;7(4):358–69.PubMedCrossRef Cotter D, Hudson L, Landau S. Evidence for orbitofrontal pathology in bipolar disorder and major depression, but not in schizophrenia. Bipolar Disord. 2005;7(4):358–69.PubMedCrossRef
225.
Zurück zum Zitat Rajkowska G, Miguel-Hidalgo JJ, Wei J, Dilley G, Pittman SD, Meltzer HY, et al. Morphometric evidence for neuronal and glial prefrontal cell pathology in major depression. Biol Psychiatry. 1999;45(9):1085–98.PubMedCrossRef Rajkowska G, Miguel-Hidalgo JJ, Wei J, Dilley G, Pittman SD, Meltzer HY, et al. Morphometric evidence for neuronal and glial prefrontal cell pathology in major depression. Biol Psychiatry. 1999;45(9):1085–98.PubMedCrossRef
226.
Zurück zum Zitat Stockmeier CA, Mahajan GJ, Konick LC, Overholser JC, Jurjus GJ, Meltzer HY, et al. Cellular changes in the postmortem hippocampus in major depression. Biol Psychiatry. 2004;56(9):640–50.PubMedPubMedCentralCrossRef Stockmeier CA, Mahajan GJ, Konick LC, Overholser JC, Jurjus GJ, Meltzer HY, et al. Cellular changes in the postmortem hippocampus in major depression. Biol Psychiatry. 2004;56(9):640–50.PubMedPubMedCentralCrossRef
227.
Zurück zum Zitat Magarinos AM, McEwen BS, Flugge G, Fuchs E. Chronic psychosocial stress causes apical dendritic atrophy of hippocampal CA3 pyramidal neurons in subordinate tree shrews. J Neurosci. 1996;16(10):3534–40.PubMedCrossRef Magarinos AM, McEwen BS, Flugge G, Fuchs E. Chronic psychosocial stress causes apical dendritic atrophy of hippocampal CA3 pyramidal neurons in subordinate tree shrews. J Neurosci. 1996;16(10):3534–40.PubMedCrossRef
228.
Zurück zum Zitat Woolley CS, Gould E, McEwen BS. Exposure to excess glucocorticoids alters dendritic morphology of adult hippocampal pyramidal neurons. Brain Res. 1990;531(1–2):225–31.PubMedCrossRef Woolley CS, Gould E, McEwen BS. Exposure to excess glucocorticoids alters dendritic morphology of adult hippocampal pyramidal neurons. Brain Res. 1990;531(1–2):225–31.PubMedCrossRef
229.
Zurück zum Zitat Watanabe Y, Gould E, McEwen BS. Stress induces atrophy of apical dendrites of hippocampal CA3 pyramidal neurons. Brain Res. 1992;588(2):341–5.PubMedCrossRef Watanabe Y, Gould E, McEwen BS. Stress induces atrophy of apical dendrites of hippocampal CA3 pyramidal neurons. Brain Res. 1992;588(2):341–5.PubMedCrossRef
230.
Zurück zum Zitat Cook SC, Wellman CL. Chronic stress alters dendritic morphology in rat medial prefrontal cortex. J Neurobiol. 2004;60(2):236–48.PubMedCrossRef Cook SC, Wellman CL. Chronic stress alters dendritic morphology in rat medial prefrontal cortex. J Neurobiol. 2004;60(2):236–48.PubMedCrossRef
231.
Zurück zum Zitat Radley JJ, Sisti HM, Hao J, Rocher AB, McCall T, Hof PR, et al. Chronic behavioral stress induces apical dendritic reorganization in pyramidal neurons of the medial prefrontal cortex. Neuroscience. 2004;125(1):1–6.PubMedCrossRef Radley JJ, Sisti HM, Hao J, Rocher AB, McCall T, Hof PR, et al. Chronic behavioral stress induces apical dendritic reorganization in pyramidal neurons of the medial prefrontal cortex. Neuroscience. 2004;125(1):1–6.PubMedCrossRef
232.
Zurück zum Zitat Wellman CL. Dendritic reorganization in pyramidal neurons in medial prefrontal cortex after chronic corticosterone administration. J Neurobiol. 2001;49(3):245–53.PubMedCrossRef Wellman CL. Dendritic reorganization in pyramidal neurons in medial prefrontal cortex after chronic corticosterone administration. J Neurobiol. 2001;49(3):245–53.PubMedCrossRef
233.
Zurück zum Zitat Goldwater DS, Pavlides C, Hunter RG, Bloss EB, Hof PR, McEwen BS, et al. Structural and functional alterations to rat medial prefrontal cortex following chronic restraint stress and recovery. Neuroscience. 2009;164(2):798–808.PubMedPubMedCentralCrossRef Goldwater DS, Pavlides C, Hunter RG, Bloss EB, Hof PR, McEwen BS, et al. Structural and functional alterations to rat medial prefrontal cortex following chronic restraint stress and recovery. Neuroscience. 2009;164(2):798–808.PubMedPubMedCentralCrossRef
234.
Zurück zum Zitat 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(9):1413–7.PubMedPubMedCentralCrossRef 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(9):1413–7.PubMedPubMedCentralCrossRef
235.
Zurück zum Zitat Bessa JM, Ferreira D, Melo I, Marques F, Cerqueira JJ, Palha JA, et al. The mood-improving actions of antidepressants do not depend on neurogenesis but are associated with neuronal remodeling. Mol Psychiatry. 2009;14(8):764–73.PubMedCrossRef Bessa JM, Ferreira D, Melo I, Marques F, Cerqueira JJ, Palha JA, et al. The mood-improving actions of antidepressants do not depend on neurogenesis but are associated with neuronal remodeling. Mol Psychiatry. 2009;14(8):764–73.PubMedCrossRef
236.
Zurück zum Zitat Cavalleri L, Merlo Pich E, Millan MJ, Chiamulera C, Kunath T, Spano PF, et al. Ketamine enhances structural plasticity in mouse mesencephalic and human iPSC-derived dopaminergic neurons via AMPAR-driven BDNF and mTOR signaling. Mol Psychiatry. 2017. https://doi.org/10.1038/mp.2017.241. (epub ahead of print). Cavalleri L, Merlo Pich E, Millan MJ, Chiamulera C, Kunath T, Spano PF, et al. Ketamine enhances structural plasticity in mouse mesencephalic and human iPSC-derived dopaminergic neurons via AMPAR-driven BDNF and mTOR signaling. Mol Psychiatry. 2017. https://​doi.​org/​10.​1038/​mp.​2017.​241. (epub ahead of print).
237.
Zurück zum Zitat Vaidya VA, Siuciak JA, Du F, Duman RS. Hippocampal mossy fiber sprouting induced by chronic electroconvulsive seizures. Neuroscience. 1999;89(1):157–66.PubMedCrossRef Vaidya VA, Siuciak JA, Du F, Duman RS. Hippocampal mossy fiber sprouting induced by chronic electroconvulsive seizures. Neuroscience. 1999;89(1):157–66.PubMedCrossRef
238.
Zurück zum Zitat Acosta-Pena E, Camacho-Abrego I, Melgarejo-Gutierrez M, Flores G, Drucker-Colin R, Garcia-Garcia F. Sleep deprivation induces differential morphological changes in the hippocampus and prefrontal cortex in young and old rats. Synapse. 2015;69(1):15–25.PubMedCrossRef Acosta-Pena E, Camacho-Abrego I, Melgarejo-Gutierrez M, Flores G, Drucker-Colin R, Garcia-Garcia F. Sleep deprivation induces differential morphological changes in the hippocampus and prefrontal cortex in young and old rats. Synapse. 2015;69(1):15–25.PubMedCrossRef
239.
Zurück zum Zitat Opal MD, Klenotich SC, Morais M, Bessa J, Winkle J, Doukas D, et al. Serotonin 2C receptor antagonists induce fast-onset antidepressant effects. Mol Psychiatry. 2014;19(10):1106–14.PubMedCrossRef Opal MD, Klenotich SC, Morais M, Bessa J, Winkle J, Doukas D, et al. Serotonin 2C receptor antagonists induce fast-onset antidepressant effects. Mol Psychiatry. 2014;19(10):1106–14.PubMedCrossRef
241.
Zurück zum Zitat Derkach VA, Oh MC, Guire ES, Soderling TR. Regulatory mechanisms of AMPA receptors in synaptic plasticity. Nat Rev Neurosci. 2007;8(2):101–13.PubMedCrossRef Derkach VA, Oh MC, Guire ES, Soderling TR. Regulatory mechanisms of AMPA receptors in synaptic plasticity. Nat Rev Neurosci. 2007;8(2):101–13.PubMedCrossRef
242.
Zurück zum Zitat Henley JM, Wilkinson KA. Synaptic AMPA receptor composition in development, plasticity and disease. Nat Rev Neurosci. 2016;17(6):337–50.PubMedCrossRef Henley JM, Wilkinson KA. Synaptic AMPA receptor composition in development, plasticity and disease. Nat Rev Neurosci. 2016;17(6):337–50.PubMedCrossRef
243.
Zurück zum Zitat Shepherd JD, Huganir RL. The cell biology of synaptic plasticity: AMPA receptor trafficking. Annu Rev Cell Dev Biol. 2007;23:613–43.PubMedCrossRef Shepherd JD, Huganir RL. The cell biology of synaptic plasticity: AMPA receptor trafficking. Annu Rev Cell Dev Biol. 2007;23:613–43.PubMedCrossRef
244.
Zurück zum Zitat Koike H, Chaki S. Requirement of AMPA receptor stimulation for the sustained antidepressant activity of ketamine and LY341495 during the forced swim test in rats. Behav Brain Res. 2014;1(271):111–5.CrossRef Koike H, Chaki S. Requirement of AMPA receptor stimulation for the sustained antidepressant activity of ketamine and LY341495 during the forced swim test in rats. Behav Brain Res. 2014;1(271):111–5.CrossRef
245.
Zurück zum Zitat Koike H, Iijima M, Chaki S. Involvement of AMPA receptor in both the rapid and sustained antidepressant-like effects of ketamine in animal models of depression. Behav Brain Res. 2011;224(1):107–11.PubMedCrossRef Koike H, Iijima M, Chaki S. Involvement of AMPA receptor in both the rapid and sustained antidepressant-like effects of ketamine in animal models of depression. Behav Brain Res. 2011;224(1):107–11.PubMedCrossRef
246.
Zurück zum Zitat Walker AK, Budac DP, Bisulco S, Lee AW, Smith RA, Beenders B, et al. NMDA receptor blockade by ketamine abrogates lipopolysaccharide-induced depressive-like behavior in C57BL/6J mice. Neuropsychopharmacology. 2013;38(9):1609–16.PubMedPubMedCentralCrossRef Walker AK, Budac DP, Bisulco S, Lee AW, Smith RA, Beenders B, et al. NMDA receptor blockade by ketamine abrogates lipopolysaccharide-induced depressive-like behavior in C57BL/6J mice. Neuropsychopharmacology. 2013;38(9):1609–16.PubMedPubMedCentralCrossRef
247.
Zurück zum Zitat Zhou W, Wang N, Yang C, Li XM, Zhou ZQ, Yang JJ. Ketamine-induced antidepressant effects are associated with AMPA receptors-mediated upregulation of mTOR and BDNF in rat hippocampus and prefrontal cortex. Eur Psychiatry. 2014;29(7):419–23.PubMedCrossRef Zhou W, Wang N, Yang C, Li XM, Zhou ZQ, Yang JJ. Ketamine-induced antidepressant effects are associated with AMPA receptors-mediated upregulation of mTOR and BDNF in rat hippocampus and prefrontal cortex. Eur Psychiatry. 2014;29(7):419–23.PubMedCrossRef
248.
Zurück zum Zitat Martin AE, Schober DA, Nikolayev A, Tolstikov VV, Anderson WH, Higgs RE, et al. Further evaluation of mechanisms associated with the antidepressant-like signature of scopolamine in mice. CNS Neurol Disord Drug Targets. 2017;16(4):492–500.PubMedCrossRef Martin AE, Schober DA, Nikolayev A, Tolstikov VV, Anderson WH, Higgs RE, et al. Further evaluation of mechanisms associated with the antidepressant-like signature of scopolamine in mice. CNS Neurol Disord Drug Targets. 2017;16(4):492–500.PubMedCrossRef
249.
Zurück zum Zitat Karasawa J, Shimazaki T, Kawashima N, Chaki S. AMPA receptor stimulation mediates the antidepressant-like effect of a group II metabotropic glutamate receptor antagonist. Brain Res. 2005;1042(1):92–8.PubMedCrossRef Karasawa J, Shimazaki T, Kawashima N, Chaki S. AMPA receptor stimulation mediates the antidepressant-like effect of a group II metabotropic glutamate receptor antagonist. Brain Res. 2005;1042(1):92–8.PubMedCrossRef
250.
Zurück zum Zitat Wolak M, Siwek A, Szewczyk B, Poleszak E, Pilc A, Popik P, et al. Involvement of NMDA and AMPA receptors in the antidepressant-like activity of antidepressant drugs in the forced swim test. Pharmacol Rep. 2013;65(4):991–7.PubMedCrossRef Wolak M, Siwek A, Szewczyk B, Poleszak E, Pilc A, Popik P, et al. Involvement of NMDA and AMPA receptors in the antidepressant-like activity of antidepressant drugs in the forced swim test. Pharmacol Rep. 2013;65(4):991–7.PubMedCrossRef
251.
Zurück zum Zitat Whittington MA, Traub RD, Kopell N, Ermentrout B, Buhl EH. Inhibition-based rhythms: experimental and mathematical observations on network dynamics. Int J Psychophysiol. 2000;38(3):315–36.PubMedCrossRef Whittington MA, Traub RD, Kopell N, Ermentrout B, Buhl EH. Inhibition-based rhythms: experimental and mathematical observations on network dynamics. Int J Psychophysiol. 2000;38(3):315–36.PubMedCrossRef
252.
Zurück zum Zitat Cunningham MO, Davies CH, Buhl EH, Kopell N, Whittington MA. Gamma oscillations induced by kainate receptor activation in the entorhinal cortex in vitro. J Neurosci. 2003;23(30):9761–9.PubMed Cunningham MO, Davies CH, Buhl EH, Kopell N, Whittington MA. Gamma oscillations induced by kainate receptor activation in the entorhinal cortex in vitro. J Neurosci. 2003;23(30):9761–9.PubMed
253.
Zurück zum Zitat 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;35(33):11694–706.PubMedPubMedCentralCrossRef 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;35(33):11694–706.PubMedPubMedCentralCrossRef
254.
Zurück zum Zitat Lazarewicz MT, Ehrlichman RS, Maxwell CR, Gandal MJ, Finkel LH, Siegel SJ. Ketamine modulates theta and gamma oscillations. J Cogn Neurosci. 2010;22(7):1452–64.PubMedCrossRef Lazarewicz MT, Ehrlichman RS, Maxwell CR, Gandal MJ, Finkel LH, Siegel SJ. Ketamine modulates theta and gamma oscillations. J Cogn Neurosci. 2010;22(7):1452–64.PubMedCrossRef
255.
Zurück zum Zitat Ahnaou A, Ver Donck L, Drinkenburg WHIM. Blockade of the metabotropic glutamate (mGluR2) modulates arousal through vigilance states transitions: evidence from sleep–wake EEG in rodents. Behav Brain Res. 2014;270:56–67.PubMedCrossRef Ahnaou A, Ver Donck L, Drinkenburg WHIM. Blockade of the metabotropic glutamate (mGluR2) modulates arousal through vigilance states transitions: evidence from sleep–wake EEG in rodents. Behav Brain Res. 2014;270:56–67.PubMedCrossRef
257.
258.
Zurück zum Zitat 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 Nat Acad Sci. 2015;112(26):8106–11.PubMedPubMedCentralCrossRef 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 Nat Acad Sci. 2015;112(26):8106–11.PubMedPubMedCentralCrossRef
259.
Zurück zum Zitat Nosyreva E, Szabla K, Autry AE, Ryazanov AG, Monteggia LM, Kavalali ET. Acute suppression of spontaneous neurotransmission drives synaptic potentiation. J Neurosci. 2013;33(16):6990–7002.PubMedPubMedCentralCrossRef Nosyreva E, Szabla K, Autry AE, Ryazanov AG, Monteggia LM, Kavalali ET. Acute suppression of spontaneous neurotransmission drives synaptic potentiation. J Neurosci. 2013;33(16):6990–7002.PubMedPubMedCentralCrossRef
260.
Zurück zum Zitat Bjorkholm C, Jardemark K, Schilstrom B, Svensson TH. Ketamine-like effects of a combination of olanzapine and fluoxetine on AMPA and NMDA receptor-mediated transmission in the medial prefrontal cortex of the rat. Eur Neuropsychopharmacol. 2015;25(10):1842–7.PubMedCrossRef Bjorkholm C, Jardemark K, Schilstrom B, Svensson TH. Ketamine-like effects of a combination of olanzapine and fluoxetine on AMPA and NMDA receptor-mediated transmission in the medial prefrontal cortex of the rat. Eur Neuropsychopharmacol. 2015;25(10):1842–7.PubMedCrossRef
261.
Zurück zum Zitat El Iskandrani KS, Oosterhof CA, El Mansari M, Blier P. Impact of subanesthetic doses of ketamine on AMPA-mediated responses in rats: an in vivo electrophysiological study on monoaminergic and glutamatergic neurons. J Psychopharmacol. 2015;29(7):792–801.PubMedPubMedCentralCrossRef El Iskandrani KS, Oosterhof CA, El Mansari M, Blier P. Impact of subanesthetic doses of ketamine on AMPA-mediated responses in rats: an in vivo electrophysiological study on monoaminergic and glutamatergic neurons. J Psychopharmacol. 2015;29(7):792–801.PubMedPubMedCentralCrossRef
262.
Zurück zum Zitat Xie M, Li C, He C, Yang L, Tan G, Yan J, et al. Short-term sleep deprivation disrupts the molecular composition of ionotropic glutamate receptors in entorhinal cortex and impairs the rat spatial reference memory. Behav Brain Res. 2016;300:70–6.PubMedCrossRef Xie M, Li C, He C, Yang L, Tan G, Yan J, et al. Short-term sleep deprivation disrupts the molecular composition of ionotropic glutamate receptors in entorhinal cortex and impairs the rat spatial reference memory. Behav Brain Res. 2016;300:70–6.PubMedCrossRef
263.
Zurück zum Zitat Xie M, Yan J, He C, Yang L, Tan G, Li C, et al. Short-term sleep deprivation impairs spatial working memory and modulates expression levels of ionotropic glutamate receptor subunits in hippocampus. Behav Brain Res. 2015;286:64–70.PubMedCrossRef Xie M, Yan J, He C, Yang L, Tan G, Li C, et al. Short-term sleep deprivation impairs spatial working memory and modulates expression levels of ionotropic glutamate receptor subunits in hippocampus. Behav Brain Res. 2015;286:64–70.PubMedCrossRef
264.
Zurück zum Zitat Zhang K, Xu T, Yuan Z, Wei Z, Yamaki VN, Huang M, et al. Essential roles of AMPA receptor GluA1 phosphorylation and presynaptic HCN channels in fast-acting antidepressant responses of ketamine. Sci Signal. 2016;9(458):ra123.PubMedPubMedCentralCrossRef Zhang K, Xu T, Yuan Z, Wei Z, Yamaki VN, Huang M, et al. Essential roles of AMPA receptor GluA1 phosphorylation and presynaptic HCN channels in fast-acting antidepressant responses of ketamine. Sci Signal. 2016;9(458):ra123.PubMedPubMedCentralCrossRef
265.
Zurück zum Zitat Lindholm JS, Autio H, Vesa L, Antila H, Lindemann L, Hoener MC, et al. The antidepressant-like effects of glutamatergic drugs ketamine and AMPA receptor potentiator LY 451646 are preserved in bdnf(+)/(−) heterozygous null mice. Neuropharmacology. 2012;62(1):391–7.PubMedCrossRef Lindholm JS, Autio H, Vesa L, Antila H, Lindemann L, Hoener MC, et al. The antidepressant-like effects of glutamatergic drugs ketamine and AMPA receptor potentiator LY 451646 are preserved in bdnf(+)/(−) heterozygous null mice. Neuropharmacology. 2012;62(1):391–7.PubMedCrossRef
266.
Zurück zum Zitat Schechter LE, Ring RH, Beyer CE, Hughes ZA, Khawaja X, Malberg JE, et al. Innovative approaches for the development of antidepressant drugs: current and future strategies. NeuroRx. 2005;2(4):590–611.PubMedPubMedCentralCrossRef Schechter LE, Ring RH, Beyer CE, Hughes ZA, Khawaja X, Malberg JE, et al. Innovative approaches for the development of antidepressant drugs: current and future strategies. NeuroRx. 2005;2(4):590–611.PubMedPubMedCentralCrossRef
267.
Zurück zum Zitat Li X, Tizzano JP, Griffey K, Clay M, Lindstrom T, Skolnick P. Antidepressant-like actions of an AMPA receptor potentiator (LY392098). Neuropharmacology. 2001;40(8):1028–33.PubMedCrossRef Li X, Tizzano JP, Griffey K, Clay M, Lindstrom T, Skolnick P. Antidepressant-like actions of an AMPA receptor potentiator (LY392098). Neuropharmacology. 2001;40(8):1028–33.PubMedCrossRef
268.
Zurück zum Zitat Akinfiresoye L, Tizabi Y. Antidepressant effects of AMPA and ketamine combination: role of hippocampal BDNF, synapsin, and mTOR. Psychopharmacology (Berl). 2013;230(2):291–8.PubMedCrossRef Akinfiresoye L, Tizabi Y. Antidepressant effects of AMPA and ketamine combination: role of hippocampal BDNF, synapsin, and mTOR. Psychopharmacology (Berl). 2013;230(2):291–8.PubMedCrossRef
269.
Zurück zum Zitat Katz LC, Shatz CJ. Synaptic activity and the construction of cortical circuits. Science. 1996;274(5290):1133–8.PubMedCrossRef Katz LC, Shatz CJ. Synaptic activity and the construction of cortical circuits. Science. 1996;274(5290):1133–8.PubMedCrossRef
270.
Zurück zum Zitat Mamounas LA, Altar CA, Blue ME, Kaplan DR, Tessarollo L, Lyons WE. BDNF promotes the regenerative sprouting, but not survival, of injured serotonergic axons in the adult rat brain. J Neurosci. 2000;20(2):771–82.PubMedCrossRef Mamounas LA, Altar CA, Blue ME, Kaplan DR, Tessarollo L, Lyons WE. BDNF promotes the regenerative sprouting, but not survival, of injured serotonergic axons in the adult rat brain. J Neurosci. 2000;20(2):771–82.PubMedCrossRef
271.
272.
Zurück zum Zitat Pattwell SS, Bath KG, Perez-Castro R, Lee FS, Chao MV, Ninan I. The BDNF Val66Met polymorphism impairs synaptic transmission and plasticity in the infralimbic medial prefrontal cortex. J Neurosci. 2012;32(7):2410–21.PubMedPubMedCentralCrossRef Pattwell SS, Bath KG, Perez-Castro R, Lee FS, Chao MV, Ninan I. The BDNF Val66Met polymorphism impairs synaptic transmission and plasticity in the infralimbic medial prefrontal cortex. J Neurosci. 2012;32(7):2410–21.PubMedPubMedCentralCrossRef
273.
Zurück zum Zitat Bocchio-Chiavetto L, Bagnardi V, Zanardini R, Molteni R, Nielsen MG, Placentino A, et al. Serum and plasma BDNF levels in major depression: a replication study and meta-analyses. World J Biol Psychiatry. 2010;11(6):763–73.PubMedCrossRef Bocchio-Chiavetto L, Bagnardi V, Zanardini R, Molteni R, Nielsen MG, Placentino A, et al. Serum and plasma BDNF levels in major depression: a replication study and meta-analyses. World J Biol Psychiatry. 2010;11(6):763–73.PubMedCrossRef
274.
Zurück zum Zitat Molendijk ML, Bus BA, Spinhoven P, Penninx BW, Kenis G, Prickaerts J, et al. Serum levels of brain-derived neurotrophic factor in major depressive disorder: state-trait issues, clinical features and pharmacological treatment. Mol Psychiatry. 2011;16(11):1088–95.PubMedCrossRef Molendijk ML, Bus BA, Spinhoven P, Penninx BW, Kenis G, Prickaerts J, et al. Serum levels of brain-derived neurotrophic factor in major depressive disorder: state-trait issues, clinical features and pharmacological treatment. Mol Psychiatry. 2011;16(11):1088–95.PubMedCrossRef
275.
Zurück zum Zitat Karlovic D, Serretti A, Jevtovic S, Vrkic N, Seric V, Peles AM. Diagnostic accuracy of serum brain derived neurotrophic factor concentration in antidepressant naive patients with first major depression episode. J Psychiatr Res. 2013;47(2):162–7.PubMedCrossRef Karlovic D, Serretti A, Jevtovic S, Vrkic N, Seric V, Peles AM. Diagnostic accuracy of serum brain derived neurotrophic factor concentration in antidepressant naive patients with first major depression episode. J Psychiatr Res. 2013;47(2):162–7.PubMedCrossRef
276.
Zurück zum Zitat Yoshida T, Ishikawa M, Niitsu T, Nakazato M, Watanabe H, Shiraishi T, et al. Decreased serum levels of mature brain-derived neurotrophic factor (BDNF), but not its precursor proBDNF, in patients with major depressive disorder. PLoS One. 2012;7(8):e42676.PubMedPubMedCentralCrossRef Yoshida T, Ishikawa M, Niitsu T, Nakazato M, Watanabe H, Shiraishi T, et al. Decreased serum levels of mature brain-derived neurotrophic factor (BDNF), but not its precursor proBDNF, in patients with major depressive disorder. PLoS One. 2012;7(8):e42676.PubMedPubMedCentralCrossRef
278.
Zurück zum Zitat Duman RS, Heninger GR, Nestler EJ. A molecular and cellular theory of depression. Arch Gen Psychiatry. 1997;54(7):597–606.PubMedCrossRef Duman RS, Heninger GR, Nestler EJ. A molecular and cellular theory of depression. Arch Gen Psychiatry. 1997;54(7):597–606.PubMedCrossRef
279.
Zurück zum Zitat Manji HK, Moore GJ, Rajkowska G, Chen G. Neuroplasticity and cellular resilience in mood disorders. Mol Psychiatry. 2000;5(6):578–93.PubMedCrossRef Manji HK, Moore GJ, Rajkowska G, Chen G. Neuroplasticity and cellular resilience in mood disorders. Mol Psychiatry. 2000;5(6):578–93.PubMedCrossRef
280.
Zurück zum Zitat Castren E, Voikar V, Rantamaki T. Role of neurotrophic factors in depression. Curr Opin Pharmacol. 2007;7(1):18–21.PubMedCrossRef Castren E, Voikar V, Rantamaki T. Role of neurotrophic factors in depression. Curr Opin Pharmacol. 2007;7(1):18–21.PubMedCrossRef
281.
Zurück zum Zitat Nibuya M, Morinobu S, Duman RS. Regulation of BDNF and trkB mRNA in rat brain by chronic electroconvulsive seizure and antidepressant drug treatments. J Neurosci. 1995;15(11):7539–47.PubMedCrossRef Nibuya M, Morinobu S, Duman RS. Regulation of BDNF and trkB mRNA in rat brain by chronic electroconvulsive seizure and antidepressant drug treatments. J Neurosci. 1995;15(11):7539–47.PubMedCrossRef
282.
Zurück zum Zitat Shimizu E, Hashimoto K, Okamura N, Koike K, Komatsu N, Kumakiri C, et al. Alterations of serum levels of brain-derived neurotrophic factor (BDNF) in depressed patients with or without antidepressants. Biol Psychiatry. 2003;54(1):70–5.PubMedCrossRef Shimizu E, Hashimoto K, Okamura N, Koike K, Komatsu N, Kumakiri C, et al. Alterations of serum levels of brain-derived neurotrophic factor (BDNF) in depressed patients with or without antidepressants. Biol Psychiatry. 2003;54(1):70–5.PubMedCrossRef
283.
Zurück zum Zitat Bocchio-Chiavetto L, Zanardini R, Bortolomasi M, Abate M, Segala M, Giacopuzzi M, et al. Electroconvulsive therapy (ECT) increases serum brain derived neurotrophic factor (BDNF) in drug resistant depressed patients. Eur Neuropsychopharmacol. 2006;16(8):620–4.PubMedCrossRef Bocchio-Chiavetto L, Zanardini R, Bortolomasi M, Abate M, Segala M, Giacopuzzi M, et al. Electroconvulsive therapy (ECT) increases serum brain derived neurotrophic factor (BDNF) in drug resistant depressed patients. Eur Neuropsychopharmacol. 2006;16(8):620–4.PubMedCrossRef
284.
Zurück zum Zitat Adachi M, Barrot M, Autry AE, Theobald D, Monteggia LM. Selective loss of brain-derived neurotrophic factor in the dentate gyrus attenuates antidepressant efficacy. Biol Psychiatry. 2008;63(7):642–9.PubMedCrossRef Adachi M, Barrot M, Autry AE, Theobald D, Monteggia LM. Selective loss of brain-derived neurotrophic factor in the dentate gyrus attenuates antidepressant efficacy. Biol Psychiatry. 2008;63(7):642–9.PubMedCrossRef
285.
Zurück zum Zitat Monteggia LM, Luikart B, Barrot M, Theobold D, Malkovska I, Nef S, et al. Brain-derived neurotrophic factor conditional knockouts show gender differences in depression-related behaviors. Biol Psychiatry. 2007;61(2):187–97.PubMedCrossRef Monteggia LM, Luikart B, Barrot M, Theobold D, Malkovska I, Nef S, et al. Brain-derived neurotrophic factor conditional knockouts show gender differences in depression-related behaviors. Biol Psychiatry. 2007;61(2):187–97.PubMedCrossRef
286.
Zurück zum Zitat Koponen E, Rantamaki T, Voikar V, Saarelainen T, MacDonald E, Castren E. Enhanced BDNF signaling is associated with an antidepressant-like behavioral response and changes in brain monoamines. Cell Mol Neurobiol. 2005;25(6):973–80.PubMedCrossRef Koponen E, Rantamaki T, Voikar V, Saarelainen T, MacDonald E, Castren E. Enhanced BDNF signaling is associated with an antidepressant-like behavioral response and changes in brain monoamines. Cell Mol Neurobiol. 2005;25(6):973–80.PubMedCrossRef
287.
Zurück zum Zitat Hoshaw BA, Malberg JE, Lucki I. Central administration of IGF-I and BDNF leads to long-lasting antidepressant-like effects. Brain Res. 2005;1037(1–2):204–8.PubMedCrossRef Hoshaw BA, Malberg JE, Lucki I. Central administration of IGF-I and BDNF leads to long-lasting antidepressant-like effects. Brain Res. 2005;1037(1–2):204–8.PubMedCrossRef
288.
Zurück zum Zitat Shirayama Y, Chen AC, Nakagawa S, Russell DS, Duman RS. Brain-derived neurotrophic factor produces antidepressant effects in behavioral models of depression. J Neurosci. 2002;22(8):3251–61.PubMedCrossRef Shirayama Y, Chen AC, Nakagawa S, Russell DS, Duman RS. Brain-derived neurotrophic factor produces antidepressant effects in behavioral models of depression. J Neurosci. 2002;22(8):3251–61.PubMedCrossRef
289.
Zurück zum Zitat Schmidt HD, Duman RS. Peripheral BDNF produces antidepressant-like effects in cellular and behavioral models. Neuropsychopharmacology. 2010;35(12):2378–91.PubMedPubMedCentralCrossRef Schmidt HD, Duman RS. Peripheral BDNF produces antidepressant-like effects in cellular and behavioral models. Neuropsychopharmacology. 2010;35(12):2378–91.PubMedPubMedCentralCrossRef
290.
Zurück zum Zitat Taliaz D, Loya A, Gersner R, Haramati S, Chen A, Zangen A. Resilience to chronic stress is mediated by hippocampal brain-derived neurotrophic factor. J Neurosci. 2011;31(12):4475–83.PubMedCrossRef Taliaz D, Loya A, Gersner R, Haramati S, Chen A, Zangen A. Resilience to chronic stress is mediated by hippocampal brain-derived neurotrophic factor. J Neurosci. 2011;31(12):4475–83.PubMedCrossRef
291.
Zurück zum Zitat Rantamaki T, Hendolin P, Kankaanpaa A, Mijatovic J, Piepponen P, Domenici E, et al. Pharmacologically diverse antidepressants rapidly activate brain-derived neurotrophic factor receptor TrkB and induce phospholipase-Cgamma signaling pathways in mouse brain. Neuropsychopharmacology. 2007;32(10):2152–62.PubMedCrossRef Rantamaki T, Hendolin P, Kankaanpaa A, Mijatovic J, Piepponen P, Domenici E, et al. Pharmacologically diverse antidepressants rapidly activate brain-derived neurotrophic factor receptor TrkB and induce phospholipase-Cgamma signaling pathways in mouse brain. Neuropsychopharmacology. 2007;32(10):2152–62.PubMedCrossRef
292.
Zurück zum Zitat Saarelainen T, Hendolin P, Lucas G, Koponen E, Sairanen M, MacDonald E, et al. Activation of the TrkB neurotrophin receptor is induced by antidepressant drugs and is required for antidepressant-induced behavioral effects. J Neurosci. 2003;23(1):349–57.PubMed Saarelainen T, Hendolin P, Lucas G, Koponen E, Sairanen M, MacDonald E, et al. Activation of the TrkB neurotrophin receptor is induced by antidepressant drugs and is required for antidepressant-induced behavioral effects. J Neurosci. 2003;23(1):349–57.PubMed
293.
Zurück zum Zitat Groves JO. Is it time to reassess the BDNF hypothesis of depression? Mol Psychiatry. 2007;12(12):1079–88.PubMedCrossRef Groves JO. Is it time to reassess the BDNF hypothesis of depression? Mol Psychiatry. 2007;12(12):1079–88.PubMedCrossRef
294.
Zurück zum Zitat Wook Koo J, Labonte B, Engmann O, Calipari ES, Juarez B, Lorsch Z, et al. Essential role of mesolimbic brain-derived neurotrophic factor in chronic social stress-induced depressive behaviors. Biol Psychiatry. 2016;80(6):469–78.PubMedCrossRef Wook Koo J, Labonte B, Engmann O, Calipari ES, Juarez B, Lorsch Z, et al. Essential role of mesolimbic brain-derived neurotrophic factor in chronic social stress-induced depressive behaviors. Biol Psychiatry. 2016;80(6):469–78.PubMedCrossRef
296.
Zurück zum Zitat Chen ZY, Jing D, Bath KG, Ieraci A, Khan T, Siao CJ, et al. Genetic variant BDNF (Val66Met) polymorphism alters anxiety-related behavior. Science. 2006;314(5796):140–3.PubMedPubMedCentralCrossRef Chen ZY, Jing D, Bath KG, Ieraci A, Khan T, Siao CJ, et al. Genetic variant BDNF (Val66Met) polymorphism alters anxiety-related behavior. Science. 2006;314(5796):140–3.PubMedPubMedCentralCrossRef
297.
Zurück zum Zitat Liu RJ, Lee FS, Li XY, Bambico F, Duman RS, Aghajanian GK. Brain-derived neurotrophic factor Val66Met allele impairs basal and ketamine-stimulated synaptogenesis in prefrontal cortex. Biol Psychiatry. 2012;71(11):996–1005.PubMedCrossRef Liu RJ, Lee FS, Li XY, Bambico F, Duman RS, Aghajanian GK. Brain-derived neurotrophic factor Val66Met allele impairs basal and ketamine-stimulated synaptogenesis in prefrontal cortex. Biol Psychiatry. 2012;71(11):996–1005.PubMedCrossRef
298.
Zurück zum Zitat Ghosal S, Bang E, Yue W, Hare BD, Lepack AE, Girgenti MJ, et al. Activity-dependent BDNF release is required for the rapid antidepressant actions of scopolamine. Biol Psychiatry. 2018;83(1):29–37.PubMedCrossRef Ghosal S, Bang E, Yue W, Hare BD, Lepack AE, Girgenti MJ, et al. Activity-dependent BDNF release is required for the rapid antidepressant actions of scopolamine. Biol Psychiatry. 2018;83(1):29–37.PubMedCrossRef
300.
Zurück zum Zitat Laje G, Lally N, Mathews D, Brutsche N, Chemerinski A, Akula N, et al. Brain-derived neurotrophic factor Val66Met polymorphism and antidepressant efficacy of ketamine in depressed patients. Biol Psychiatry. 2012;72(11):e27–8.PubMedPubMedCentralCrossRef Laje G, Lally N, Mathews D, Brutsche N, Chemerinski A, Akula N, et al. Brain-derived neurotrophic factor Val66Met polymorphism and antidepressant efficacy of ketamine in depressed patients. Biol Psychiatry. 2012;72(11):e27–8.PubMedPubMedCentralCrossRef
301.
Zurück zum Zitat Garcia LS, Comim CM, Valvassori SS, Reus GZ, Barbosa LM, Andreazza AC, et al. Acute administration of ketamine induces antidepressant-like effects in the forced swimming test and increases BDNF levels in the rat hippocampus. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32(1):140–4.PubMedCrossRef Garcia LS, Comim CM, Valvassori SS, Reus GZ, Barbosa LM, Andreazza AC, et al. Acute administration of ketamine induces antidepressant-like effects in the forced swimming test and increases BDNF levels in the rat hippocampus. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32(1):140–4.PubMedCrossRef
302.
Zurück zum Zitat Newton SS, Collier EF, Hunsberger J, Adams D, Terwilliger R, Selvanayagam E, et al. Gene profile of electroconvulsive seizures: induction of neurotrophic and angiogenic factors. J Neurosci. 2003;23(34):10841–51.PubMed Newton SS, Collier EF, Hunsberger J, Adams D, Terwilliger R, Selvanayagam E, et al. Gene profile of electroconvulsive seizures: induction of neurotrophic and angiogenic factors. J Neurosci. 2003;23(34):10841–51.PubMed
303.
Zurück zum Zitat Conti B, Maier R, Barr AM, Morale MC, Lu X, Sanna PP, et al. Region-specific transcriptional changes following the three antidepressant treatments electro convulsive therapy, sleep deprivation and fluoxetine. Mol Psychiatry. 2007;12(2):167–89.PubMedCrossRef Conti B, Maier R, Barr AM, Morale MC, Lu X, Sanna PP, et al. Region-specific transcriptional changes following the three antidepressant treatments electro convulsive therapy, sleep deprivation and fluoxetine. Mol Psychiatry. 2007;12(2):167–89.PubMedCrossRef
304.
Zurück zum Zitat Altar CA, Whitehead RE, Chen R, Wortwein G, Madsen TM. Effects of electroconvulsive seizures and antidepressant drugs on brain-derived neurotrophic factor protein in rat brain. Biol Psychiatry. 2003;54(7):703–9.PubMedCrossRef Altar CA, Whitehead RE, Chen R, Wortwein G, Madsen TM. Effects of electroconvulsive seizures and antidepressant drugs on brain-derived neurotrophic factor protein in rat brain. Biol Psychiatry. 2003;54(7):703–9.PubMedCrossRef
305.
Zurück zum Zitat Sartorius A, Hellweg R, Litzke J, Vogt M, Dormann C, Vollmayr B, et al. Correlations and discrepancies between serum and brain tissue levels of neurotrophins after electroconvulsive treatment in rats. Pharmacopsychiatry. 2009;42(6):270–6.PubMedCrossRef Sartorius A, Hellweg R, Litzke J, Vogt M, Dormann C, Vollmayr B, et al. Correlations and discrepancies between serum and brain tissue levels of neurotrophins after electroconvulsive treatment in rats. Pharmacopsychiatry. 2009;42(6):270–6.PubMedCrossRef
306.
Zurück zum Zitat Balu DT, Hoshaw BA, Malberg JE, Rosenzweig-Lipson S, Schechter LE, Lucki I. Differential regulation of central BDNF protein levels by antidepressant and non-antidepressant drug treatments. Brain Res. 2008;23(1211):37–43.CrossRef Balu DT, Hoshaw BA, Malberg JE, Rosenzweig-Lipson S, Schechter LE, Lucki I. Differential regulation of central BDNF protein levels by antidepressant and non-antidepressant drug treatments. Brain Res. 2008;23(1211):37–43.CrossRef
307.
Zurück zum Zitat Li B, Suemaru K, Cui R, Araki H. Repeated electroconvulsive stimuli have long-lasting effects on hippocampal BDNF and decrease immobility time in the rat forced swim test. Life Sci. 2007;80(16):1539–43.PubMedCrossRef Li B, Suemaru K, Cui R, Araki H. Repeated electroconvulsive stimuli have long-lasting effects on hippocampal BDNF and decrease immobility time in the rat forced swim test. Life Sci. 2007;80(16):1539–43.PubMedCrossRef
308.
Zurück zum Zitat Angelucci F, Aloe L, Jimenez-Vasquez P, Mathe AA. Electroconvulsive stimuli alter the regional concentrations of nerve growth factor, brain-derived neurotrophic factor, and glial cell line-derived neurotrophic factor in adult rat brain. J ECT. 2002;18(3):138–43.PubMedCrossRef Angelucci F, Aloe L, Jimenez-Vasquez P, Mathe AA. Electroconvulsive stimuli alter the regional concentrations of nerve growth factor, brain-derived neurotrophic factor, and glial cell line-derived neurotrophic factor in adult rat brain. J ECT. 2002;18(3):138–43.PubMedCrossRef
309.
Zurück zum Zitat Chen AC, Shin KH, Duman RS, Sanacora G. ECS-Induced mossy fiber sprouting and BDNF expression are attenuated by ketamine pretreatment. J ECT. 2001;17(1):27–32.PubMedCrossRef Chen AC, Shin KH, Duman RS, Sanacora G. ECS-Induced mossy fiber sprouting and BDNF expression are attenuated by ketamine pretreatment. J ECT. 2001;17(1):27–32.PubMedCrossRef
310.
Zurück zum Zitat Luo J, Min S, Wei K, Cao J, Wang B, Li P, et al. Behavioral and molecular responses to electroconvulsive shock differ between genetic and environmental rat models of depression. Psychiatry Res. 2015;226(2–3):451–60.PubMedCrossRef Luo J, Min S, Wei K, Cao J, Wang B, Li P, et al. Behavioral and molecular responses to electroconvulsive shock differ between genetic and environmental rat models of depression. Psychiatry Res. 2015;226(2–3):451–60.PubMedCrossRef
311.
Zurück zum Zitat Gersner R, Toth E, Isserles M, Zangen A. Site-specific antidepressant effects of repeated subconvulsive electrical stimulation: potential role of brain-derived neurotrophic factor. Biol Psychiatry. 2010;67(2):125–32.PubMedCrossRef Gersner R, Toth E, Isserles M, Zangen A. Site-specific antidepressant effects of repeated subconvulsive electrical stimulation: potential role of brain-derived neurotrophic factor. Biol Psychiatry. 2010;67(2):125–32.PubMedCrossRef
312.
Zurück zum Zitat Vollmayr B, Faust H, Lewicka S, Henn FA. Brain-derived-neurotrophic-factor (BDNF) stress response in rats bred for learned helplessness. Mol Psychiatry. 2001;6(4):471–4.PubMedCrossRef Vollmayr B, Faust H, Lewicka S, Henn FA. Brain-derived-neurotrophic-factor (BDNF) stress response in rats bred for learned helplessness. Mol Psychiatry. 2001;6(4):471–4.PubMedCrossRef
313.
Zurück zum Zitat Fujihara H, Sei H, Morita Y, Ueta Y, Morita K. Short-term sleep disturbance enhances brain-derived neurotrophic factor gene expression in rat hippocampus by acting as internal stressor. J Mol Neurosci. 2003;21(3):223–32.PubMedCrossRef Fujihara H, Sei H, Morita Y, Ueta Y, Morita K. Short-term sleep disturbance enhances brain-derived neurotrophic factor gene expression in rat hippocampus by acting as internal stressor. J Mol Neurosci. 2003;21(3):223–32.PubMedCrossRef
314.
Zurück zum Zitat Guo L, Guo Z, Luo X, Liang R, Yang S, Ren H, et al. Phosphodiesterase 10A inhibition attenuates sleep deprivation-induced deficits in long-term fear memory. Neurosci Lett. 2016;635:44–50.PubMedCrossRef Guo L, Guo Z, Luo X, Liang R, Yang S, Ren H, et al. Phosphodiesterase 10A inhibition attenuates sleep deprivation-induced deficits in long-term fear memory. Neurosci Lett. 2016;635:44–50.PubMedCrossRef
315.
Zurück zum Zitat Guzman-Marin R, Ying Z, Suntsova N, Methippara M, Bashir T, Szymusiak R, et al. Suppression of hippocampal plasticity-related gene expression by sleep deprivation in rats. J Physiol. 2006;575(Pt 3):807–19.PubMedPubMedCentralCrossRef Guzman-Marin R, Ying Z, Suntsova N, Methippara M, Bashir T, Szymusiak R, et al. Suppression of hippocampal plasticity-related gene expression by sleep deprivation in rats. J Physiol. 2006;575(Pt 3):807–19.PubMedPubMedCentralCrossRef
316.
Zurück zum Zitat Jiang Y, Zhu J. Effects of sleep deprivation on behaviors and abnormal hippocampal BDNF/miR-10B expression in rats with chronic stress depression. Int J Clin Exp Pathol. 2015;8(1):586–93.PubMedPubMedCentral Jiang Y, Zhu J. Effects of sleep deprivation on behaviors and abnormal hippocampal BDNF/miR-10B expression in rats with chronic stress depression. Int J Clin Exp Pathol. 2015;8(1):586–93.PubMedPubMedCentral
317.
Zurück zum Zitat Konar A, Shah N, Singh R, Saxena N, Kaul SC, Wadhwa R, et al. Protective role of Ashwagandha leaf extract and its component withanone on scopolamine-induced changes in the brain and brain-derived cells. PLoS One. 2011;6(11):e27265.PubMedPubMedCentralCrossRef Konar A, Shah N, Singh R, Saxena N, Kaul SC, Wadhwa R, et al. Protective role of Ashwagandha leaf extract and its component withanone on scopolamine-induced changes in the brain and brain-derived cells. PLoS One. 2011;6(11):e27265.PubMedPubMedCentralCrossRef
318.
Zurück zum Zitat Lee B, Sur B, Shim J, Hahm DH, Lee H. Acupuncture stimulation improves scopolamine-induced cognitive impairment via activation of cholinergic system and regulation of BDNF and CREB expressions in rats. BMC Complement Altern Med. 2014;17(14):338.CrossRef Lee B, Sur B, Shim J, Hahm DH, Lee H. Acupuncture stimulation improves scopolamine-induced cognitive impairment via activation of cholinergic system and regulation of BDNF and CREB expressions in rats. BMC Complement Altern Med. 2014;17(14):338.CrossRef
319.
Zurück zum Zitat Chen W, Cheng X, Chen J, Yi X, Nie D, Sun X, et al. Lycium barbarum polysaccharides prevent memory and neurogenesis impairments in scopolamine-treated rats. PLoS One. 2014;9(2):e88076.PubMedPubMedCentralCrossRef Chen W, Cheng X, Chen J, Yi X, Nie D, Sun X, et al. Lycium barbarum polysaccharides prevent memory and neurogenesis impairments in scopolamine-treated rats. PLoS One. 2014;9(2):e88076.PubMedPubMedCentralCrossRef
320.
Zurück zum Zitat Kotani S, Yamauchi T, Teramoto T, Ogura H. Donepezil, an acetylcholinesterase inhibitor, enhances adult hippocampal neurogenesis. Chem Biol Interact. 2008;175(1–3):227–30.PubMedCrossRef Kotani S, Yamauchi T, Teramoto T, Ogura H. Donepezil, an acetylcholinesterase inhibitor, enhances adult hippocampal neurogenesis. Chem Biol Interact. 2008;175(1–3):227–30.PubMedCrossRef
321.
Zurück zum Zitat Shi Z, Chen L, Li S, Chen S, Sun X, Sun L, et al. Chronic scopolamine-injection-induced cognitive deficit on reward-directed instrumental learning in rat is associated with CREB signaling activity in the cerebral cortex and dorsal hippocampus. Psychopharmacology (Berl). 2013;230(2):245–60.PubMedCrossRef Shi Z, Chen L, Li S, Chen S, Sun X, Sun L, et al. Chronic scopolamine-injection-induced cognitive deficit on reward-directed instrumental learning in rat is associated with CREB signaling activity in the cerebral cortex and dorsal hippocampus. Psychopharmacology (Berl). 2013;230(2):245–60.PubMedCrossRef
322.
Zurück zum Zitat Heo YM, Shin MS, Kim SH, Kim TW, Baek SB, Baek SS. Treadmill exercise ameliorates disturbance of spatial learning ability in scopolamine-induced amnesia rats. J Exerc Rehabil. 2014;10(3):155–61.PubMedPubMedCentralCrossRef Heo YM, Shin MS, Kim SH, Kim TW, Baek SB, Baek SS. Treadmill exercise ameliorates disturbance of spatial learning ability in scopolamine-induced amnesia rats. J Exerc Rehabil. 2014;10(3):155–61.PubMedPubMedCentralCrossRef
323.
Zurück zum Zitat Weeks HR 3rd, Tadler SC, Smith KW, Iacob E, Saccoman M, White AT, et al. Antidepressant and neurocognitive effects of isoflurane anesthesia versus electroconvulsive therapy in refractory depression. PLoS One. 2013;8(7):e69809.PubMedPubMedCentralCrossRef Weeks HR 3rd, Tadler SC, Smith KW, Iacob E, Saccoman M, White AT, et al. Antidepressant and neurocognitive effects of isoflurane anesthesia versus electroconvulsive therapy in refractory depression. PLoS One. 2013;8(7):e69809.PubMedPubMedCentralCrossRef
324.
Zurück zum Zitat Langer G, Neumark J, Koinig G, Graf M, Schonbeck G. Rapid psychotherapeutic effects of anesthesia with isoflurane (ES narcotherapy) in treatment-refractory depressed patients. Neuropsychobiology. 1985;14(3):118–20.PubMedCrossRef Langer G, Neumark J, Koinig G, Graf M, Schonbeck G. Rapid psychotherapeutic effects of anesthesia with isoflurane (ES narcotherapy) in treatment-refractory depressed patients. Neuropsychobiology. 1985;14(3):118–20.PubMedCrossRef
325.
Zurück zum Zitat Langer G, Karazman R, Neumark J, Saletu B, Schonbeck G, Grunberger J, et al. Isoflurane narcotherapy in depressive patients refractory to conventional antidepressant drug treatment. A double-blind comparison with electroconvulsive treatment. Neuropsychobiology. 1995;31(4):182–94.PubMedCrossRef Langer G, Karazman R, Neumark J, Saletu B, Schonbeck G, Grunberger J, et al. Isoflurane narcotherapy in depressive patients refractory to conventional antidepressant drug treatment. A double-blind comparison with electroconvulsive treatment. Neuropsychobiology. 1995;31(4):182–94.PubMedCrossRef
326.
Zurück zum Zitat Antila H, Ryazantseva M, Popova D, Sipila P, Guirado R, Kohtala S, et al. Isoflurane produces antidepressant effects and induces TrkB signaling in rodents. Sci Rep. 2017;7(1):7811.PubMedPubMedCentralCrossRef Antila H, Ryazantseva M, Popova D, Sipila P, Guirado R, Kohtala S, et al. Isoflurane produces antidepressant effects and induces TrkB signaling in rodents. Sci Rep. 2017;7(1):7811.PubMedPubMedCentralCrossRef
327.
Zurück zum Zitat Taha E, Gildish I, Gal-Ben-Ari S, Rosenblum K. The role of eEF2 pathway in learning and synaptic plasticity. Neurobiol Learn Mem. 2013;105:100–6.PubMedCrossRef Taha E, Gildish I, Gal-Ben-Ari S, Rosenblum K. The role of eEF2 pathway in learning and synaptic plasticity. Neurobiol Learn Mem. 2013;105:100–6.PubMedCrossRef
328.
Zurück zum Zitat Chotiner JK, Khorasani H, Nairn AC, O’Dell TJ, Watson JB. Adenylyl cyclase-dependent form of chemical long-term potentiation triggers translational regulation at the elongation step. Neuroscience. 2003;116(3):743–52.PubMedCrossRef Chotiner JK, Khorasani H, Nairn AC, O’Dell TJ, Watson JB. Adenylyl cyclase-dependent form of chemical long-term potentiation triggers translational regulation at the elongation step. Neuroscience. 2003;116(3):743–52.PubMedCrossRef
329.
Zurück zum Zitat Park S, Park JM, Kim S, Kim JA, Shepherd JD, Smith-Hicks CL, et al. Elongation factor 2 and fragile X mental retardation protein control the dynamic translation of Arc/Arg3.1 essential for mGluR-LTD. Neuron. 2008;59(1):70–83.PubMedPubMedCentralCrossRef Park S, Park JM, Kim S, Kim JA, Shepherd JD, Smith-Hicks CL, et al. Elongation factor 2 and fragile X mental retardation protein control the dynamic translation of Arc/Arg3.1 essential for mGluR-LTD. Neuron. 2008;59(1):70–83.PubMedPubMedCentralCrossRef
330.
Zurück zum Zitat Lu Y, Wang C, Xue Z, Li C, Zhang J, Zhao X, et al. PI3K/AKT/mTOR signaling-mediated neuropeptide VGF in the hippocampus of mice is involved in the rapid onset antidepressant-like effects of GLYX-13. Int J Neuropsychopharmacol. 2014;18(5). https://doi.org/10.1093/ijnp/pyu110. Lu Y, Wang C, Xue Z, Li C, Zhang J, Zhao X, et al. PI3K/AKT/mTOR signaling-mediated neuropeptide VGF in the hippocampus of mice is involved in the rapid onset antidepressant-like effects of GLYX-13. Int J Neuropsychopharmacol. 2014;18(5). https://​doi.​org/​10.​1093/​ijnp/​pyu110.
331.
Zurück zum Zitat Hizli AA, Chi Y, Swanger J, Carter JH, Liao Y, Welcker M, et al. Phosphorylation of eukaryotic elongation factor 2 (eEF2) by cyclin A-cyclin-dependent kinase 2 regulates its inhibition by eEF2 kinase. Mol Cell Biol. 2013;33(3):596–604.PubMedPubMedCentralCrossRef Hizli AA, Chi Y, Swanger J, Carter JH, Liao Y, Welcker M, et al. Phosphorylation of eukaryotic elongation factor 2 (eEF2) by cyclin A-cyclin-dependent kinase 2 regulates its inhibition by eEF2 kinase. Mol Cell Biol. 2013;33(3):596–604.PubMedPubMedCentralCrossRef
332.
Zurück zum Zitat Knebel A, Morrice N, Cohen P. A novel method to identify protein kinase substrates: eEF2 kinase is phosphorylated and inhibited by SAPK4/p38delta. EMBO J. 2001;20(16):4360–9.PubMedPubMedCentralCrossRef Knebel A, Morrice N, Cohen P. A novel method to identify protein kinase substrates: eEF2 kinase is phosphorylated and inhibited by SAPK4/p38delta. EMBO J. 2001;20(16):4360–9.PubMedPubMedCentralCrossRef
333.
Zurück zum Zitat Redpath NT, Foulstone EJ, Proud CG. Regulation of translation elongation factor-2 by insulin via a rapamycin-sensitive signalling pathway. EMBO J. 1996;15(9):2291–7.PubMedPubMedCentral Redpath NT, Foulstone EJ, Proud CG. Regulation of translation elongation factor-2 by insulin via a rapamycin-sensitive signalling pathway. EMBO J. 1996;15(9):2291–7.PubMedPubMedCentral
334.
Zurück zum Zitat Wang X, Li W, Williams M, Terada N, Alessi DR, Proud CG. Regulation of elongation factor 2 kinase by p90(RSK1) and p70 S6 kinase. EMBO J. 2001;20(16):4370–9.PubMedPubMedCentralCrossRef Wang X, Li W, Williams M, Terada N, Alessi DR, Proud CG. Regulation of elongation factor 2 kinase by p90(RSK1) and p70 S6 kinase. EMBO J. 2001;20(16):4370–9.PubMedPubMedCentralCrossRef
335.
Zurück zum Zitat Grønli J, Dagestad G, Milde AM, Murison R, Bramham CR. Post-transcriptional effects and interactions between chronic mild stress and acute sleep deprivation: regulation of translation factor and cytoplasmic polyadenylation element-binding protein phosphorylation. Behav Brain Res. 2012;235(2):251–62.PubMedCrossRef Grønli J, Dagestad G, Milde AM, Murison R, Bramham CR. Post-transcriptional effects and interactions between chronic mild stress and acute sleep deprivation: regulation of translation factor and cytoplasmic polyadenylation element-binding protein phosphorylation. Behav Brain Res. 2012;235(2):251–62.PubMedCrossRef
337.
Zurück zum Zitat Yoshii A, Constantine-Paton M. Post-synaptic BDNF-TrkB signaling in synapse maturation, plasticity and disease. Dev Neurobiol. 2010;70(5):304–22.PubMedPubMedCentral Yoshii A, Constantine-Paton M. Post-synaptic BDNF-TrkB signaling in synapse maturation, plasticity and disease. Dev Neurobiol. 2010;70(5):304–22.PubMedPubMedCentral
338.
Zurück zum Zitat Duman RS, Li N, Liu RJ, Duric V, Aghajanian G. Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology. 2012;62(1):35–41.PubMedCrossRef Duman RS, Li N, Liu RJ, Duric V, Aghajanian G. Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology. 2012;62(1):35–41.PubMedCrossRef
339.
340.
Zurück zum Zitat Hoeffer CA, Klann E. mTOR signaling: at the crossroads of plasticity, memory and disease. Trends Neurosci. 2010;33(2):67–75.PubMedCrossRef Hoeffer CA, Klann E. mTOR signaling: at the crossroads of plasticity, memory and disease. Trends Neurosci. 2010;33(2):67–75.PubMedCrossRef
341.
Zurück zum Zitat Paul RK, Singh NS, Khadeer M, Moaddel R, Sanghvi M, Green CE, et al. (R,S)-Ketamine metabolites (R,S)-norketamine and (2S,6S)-hydroxynorketamine increase the mammalian target of rapamycin (mTOR) function. Anesthesiology. 2014;121(1):149–59.PubMedPubMedCentralCrossRef Paul RK, Singh NS, Khadeer M, Moaddel R, Sanghvi M, Green CE, et al. (R,S)-Ketamine metabolites (R,S)-norketamine and (2S,6S)-hydroxynorketamine increase the mammalian target of rapamycin (mTOR) function. Anesthesiology. 2014;121(1):149–59.PubMedPubMedCentralCrossRef
342.
Zurück zum Zitat Miller OH, Yang L, Wang CC, Hargroder EA, Zhang Y, Delpire E, et al. GluN2B-containing NMDA receptors regulate depression-like behavior and are critical for the rapid antidepressant actions of ketamine. Elife. 2014;23(3):e03581. Miller OH, Yang L, Wang CC, Hargroder EA, Zhang Y, Delpire E, et al. GluN2B-containing NMDA receptors regulate depression-like behavior and are critical for the rapid antidepressant actions of ketamine. Elife. 2014;23(3):e03581.
343.
Zurück zum Zitat Yang C, Hu YM, Zhou ZQ, Zhang GF, Yang JJ. Acute administration of ketamine in rats increases hippocampal BDNF and mTOR levels during forced swimming test. Ups J Med Sci. 2013;118(1):3–8.PubMedPubMedCentralCrossRef Yang C, Hu YM, Zhou ZQ, Zhang GF, Yang JJ. Acute administration of ketamine in rats increases hippocampal BDNF and mTOR levels during forced swimming test. Ups J Med Sci. 2013;118(1):3–8.PubMedPubMedCentralCrossRef
344.
Zurück zum Zitat Zhang K, Yamaki VN, Wei Z, Zheng Y, Cai X. Differential regulation of GluA1 expression by ketamine and memantine. Behav Brain Res. 2017;1(316):152–9.CrossRef Zhang K, Yamaki VN, Wei Z, Zheng Y, Cai X. Differential regulation of GluA1 expression by ketamine and memantine. Behav Brain Res. 2017;1(316):152–9.CrossRef
345.
Zurück zum Zitat Holubova K, Kleteckova L, Skurlova M, Ricny J, Stuchlik A, Vales K. Rapamycin blocks the antidepressant effect of ketamine in task-dependent manner. Psychopharmacology (Berl). 2016;233(11):2077–97.PubMedCrossRef Holubova K, Kleteckova L, Skurlova M, Ricny J, Stuchlik A, Vales K. Rapamycin blocks the antidepressant effect of ketamine in task-dependent manner. Psychopharmacology (Berl). 2016;233(11):2077–97.PubMedCrossRef
346.
Zurück zum Zitat Beurel E, Grieco SF, Jope RS. Glycogen synthase kinase-3 (GSK3): regulation, actions, and diseases. Pharmacol Ther. 2015;148:114–31.PubMedCrossRef Beurel E, Grieco SF, Jope RS. Glycogen synthase kinase-3 (GSK3): regulation, actions, and diseases. Pharmacol Ther. 2015;148:114–31.PubMedCrossRef
347.
348.
Zurück zum Zitat Zhou W, Dong L, Wang N, Shi JY, Yang JJ, Zuo ZY, et al. Akt mediates GSK-3beta phosphorylation in the rat prefrontal cortex during the process of ketamine exerting rapid antidepressant actions. Neuroimmunomodulation. 2014;21(4):183–8.PubMedCrossRef Zhou W, Dong L, Wang N, Shi JY, Yang JJ, Zuo ZY, et al. Akt mediates GSK-3beta phosphorylation in the rat prefrontal cortex during the process of ketamine exerting rapid antidepressant actions. Neuroimmunomodulation. 2014;21(4):183–8.PubMedCrossRef
349.
Zurück zum Zitat Beurel E, Grieco SF, Amadei C, Downey K, Jope RS. Ketamine-induced inhibition of glycogen synthase kinase-3 contributes to the augmentation of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor signaling. Bipolar Disord. 2016;18(6):473–80.PubMedPubMedCentralCrossRef Beurel E, Grieco SF, Amadei C, Downey K, Jope RS. Ketamine-induced inhibition of glycogen synthase kinase-3 contributes to the augmentation of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor signaling. Bipolar Disord. 2016;18(6):473–80.PubMedPubMedCentralCrossRef
350.
Zurück zum Zitat Basar K, Eren-Kocak E, Ozdemir H, Ertugrul A. Effects of acute and chronic electroconvulsive shocks on glycogen synthase kinase 3β level and phosphorylation in mice. J ECT. 2013;29(4):265–70.PubMedCrossRef Basar K, Eren-Kocak E, Ozdemir H, Ertugrul A. Effects of acute and chronic electroconvulsive shocks on glycogen synthase kinase 3β level and phosphorylation in mice. J ECT. 2013;29(4):265–70.PubMedCrossRef
351.
Zurück zum Zitat Roh MS, Kang UG, Shin SY, Lee YH, Jung HY, Juhnn YS, et al. Biphasic changes in the Ser-9 phosphorylation of glycogen synthase kinase-3beta after electroconvulsive shock in the rat brain. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27(1):1–5.PubMedCrossRef Roh MS, Kang UG, Shin SY, Lee YH, Jung HY, Juhnn YS, et al. Biphasic changes in the Ser-9 phosphorylation of glycogen synthase kinase-3beta after electroconvulsive shock in the rat brain. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27(1):1–5.PubMedCrossRef
352.
Zurück zum Zitat Kang UG, Roh MS, Jung JR, Shin SY, Lee YH, Park JB, et al. Activation of protein kinase B (Akt) signaling after electroconvulsive shock in the rat hippocampus. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28(1):41–4.PubMedCrossRef Kang UG, Roh MS, Jung JR, Shin SY, Lee YH, Park JB, et al. Activation of protein kinase B (Akt) signaling after electroconvulsive shock in the rat hippocampus. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28(1):41–4.PubMedCrossRef
353.
Zurück zum Zitat Daly EJ, Singh JB, Fedgchin M, Cooper K, Lim P, Shelton RC, et al. Efficacy and safety of intranasal esketamine adjunctive to oral antidepressant therapy in treatment-resistant depression: A randomized clinical trial. JAMA Psychiatry. 2018; 75(2):139–48PubMedCrossRef Daly EJ, Singh JB, Fedgchin M, Cooper K, Lim P, Shelton RC, et al. Efficacy and safety of intranasal esketamine adjunctive to oral antidepressant therapy in treatment-resistant depression: A randomized clinical trial. JAMA Psychiatry. 2018; 75(2):139–48PubMedCrossRef
354.
Zurück zum Zitat Zanos P, Moaddel R, Morris PJ, Wainer IW, Albuquerque EX, Thompson SM, et al. Reply to: Antidepressant actions of ketamine versus hydroxynorketamine. Biol Psychiatry. 2017;81(8):e69–71.PubMedCrossRef Zanos P, Moaddel R, Morris PJ, Wainer IW, Albuquerque EX, Thompson SM, et al. Reply to: Antidepressant actions of ketamine versus hydroxynorketamine. Biol Psychiatry. 2017;81(8):e69–71.PubMedCrossRef
356.
Zurück zum Zitat Wohleb ES, Gerhard D, Thomas A, Duman RS. Molecular and cellular mechanisms of rapid-acting antidepressants ketamine and scopolamine. Curr Neuropharmacol. 2017;15(1):11–20.PubMedPubMedCentralCrossRef Wohleb ES, Gerhard D, Thomas A, Duman RS. Molecular and cellular mechanisms of rapid-acting antidepressants ketamine and scopolamine. Curr Neuropharmacol. 2017;15(1):11–20.PubMedPubMedCentralCrossRef
357.
Zurück zum Zitat Wilkinson ST, Toprak M, Turner MS, Levine SP, Katz RB, Sanacora G. A survey of the clinical, off-label use of ketamine as a treatment for psychiatric disorders. Am J Psychiatry. 2017;174(7):695–6.PubMedPubMedCentralCrossRef Wilkinson ST, Toprak M, Turner MS, Levine SP, Katz RB, Sanacora G. A survey of the clinical, off-label use of ketamine as a treatment for psychiatric disorders. Am J Psychiatry. 2017;174(7):695–6.PubMedPubMedCentralCrossRef
358.
Zurück zum Zitat Sos P, Klirova M, Novak T, Kohutova B, Horacek J, Palenicek T. Relationship of ketamine’s antidepressant and psychotomimetic effects in unipolar depression. Neuro Endocrinol Lett. 2013;34(4):287–93.PubMed Sos P, Klirova M, Novak T, Kohutova B, Horacek J, Palenicek T. Relationship of ketamine’s antidepressant and psychotomimetic effects in unipolar depression. Neuro Endocrinol Lett. 2013;34(4):287–93.PubMed
359.
Zurück zum Zitat Hu YD, Xiang YT, Fang JX, Zu S, Sha S, Shi H, et al. Single i.v. ketamine augmentation of newly initiated escitalopram for major depression: results from a randomized, placebo-controlled 4-week study. Psychol Med. 2016;46(3):623–35.PubMedCrossRef Hu YD, Xiang YT, Fang JX, Zu S, Sha S, Shi H, et al. Single i.v. ketamine augmentation of newly initiated escitalopram for major depression: results from a randomized, placebo-controlled 4-week study. Psychol Med. 2016;46(3):623–35.PubMedCrossRef
360.
Zurück zum Zitat 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(8):816–26.PubMedCrossRef 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(8):816–26.PubMedCrossRef
361.
Zurück zum Zitat Lieberman JA, Papadakis K, Csernansky J, Litman R, Volavka J, Jia XD, et al. A randomized, placebo-controlled study of memantine as adjunctive treatment in patients with schizophrenia. Neuropsychopharmacology. 2009;34(5):1322–9.PubMedCrossRef Lieberman JA, Papadakis K, Csernansky J, Litman R, Volavka J, Jia XD, et al. A randomized, placebo-controlled study of memantine as adjunctive treatment in patients with schizophrenia. Neuropsychopharmacology. 2009;34(5):1322–9.PubMedCrossRef
362.
Zurück zum Zitat Smith EG, Deligiannidis KM, Ulbricht CM, Landolin CS, Patel JK, Rothschild AJ. Antidepressant augmentation using the NMDA-antagonist memantine: a randomized, double-blind, placebo-controlled trial. J Clin Psychiatry. 2013;74(10):966–73.PubMedPubMedCentralCrossRef Smith EG, Deligiannidis KM, Ulbricht CM, Landolin CS, Patel JK, Rothschild AJ. Antidepressant augmentation using the NMDA-antagonist memantine: a randomized, double-blind, placebo-controlled trial. J Clin Psychiatry. 2013;74(10):966–73.PubMedPubMedCentralCrossRef
Metadaten
Titel
Convergent Mechanisms Underlying Rapid Antidepressant Action
verfasst von
Panos Zanos
Scott M. Thompson
Ronald S. Duman
Carlos A. Zarate Jr.
Todd D. Gould
Publikationsdatum
01.03.2018
Verlag
Springer International Publishing
Erschienen in
CNS Drugs / Ausgabe 3/2018
Print ISSN: 1172-7047
Elektronische ISSN: 1179-1934
DOI
https://doi.org/10.1007/s40263-018-0492-x

Weitere Artikel der Ausgabe 3/2018

CNS Drugs 3/2018 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Viertel reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Bluttest erkennt Parkinson schon zehn Jahre vor der Diagnose

10.05.2024 Parkinson-Krankheit Nachrichten

Ein Bluttest kann abnorm aggregiertes Alpha-Synuclein bei einigen Menschen schon zehn Jahre vor Beginn der motorischen Parkinsonsymptome nachweisen. Mit einem solchen Test lassen sich möglicherweise Prodromalstadien erfassen und die Betroffenen früher behandeln.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Wartezeit nicht kürzer, aber Arbeit flexibler

Psychotherapie Medizin aktuell

Fünf Jahren nach der Neugestaltung der Psychotherapie-Richtlinie wurden jetzt die Effekte der vorgenommenen Änderungen ausgewertet. Das Hauptziel der Novellierung war eine kürzere Wartezeit auf Therapieplätze. Dieses Ziel wurde nicht erreicht, es gab jedoch positive Auswirkungen auf andere Bereiche.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.