Skip to main content
Log in

Targeting the Glutamatergic System to Treat Major Depressive Disorder

Rationale and Progress to Date

  • Leading Article
  • Published:
Drugs Aims and scope Submit manuscript

Abstract

Major depressive disorder (MDD) is a severe, debilitating medical illness that affects millions of individuals worldwide. The young age of onset and chronicity of the disorder has a significant impact on the long-term disability that affected individuals face. Most existing treatments have focused on the ‘monoamine hypothesis’ for rational design of compounds. However, patients continue to experience low remission rates, residual subsyndromal symptoms, relapses and overall functional impairment.

In this context, growing evidence suggests that the glutamatergic system is uniquely central to the neurobiology and treatment of MDD. Here, we review data supporting the involvement of the glutamatergic system in the pathophysiology of MDD, and discuss the efficacy of glutamatergic agents as novel therapeutics. Preliminary clinical evidence has been promising, particularly with regard to the N-methyl-D-aspartate (NMDA) antagonist ketamine as a ‘proof-of-concept’ agent. The review also highlights potential molecular and inflammatory mechanisms that may contribute to the rapid antidepressant response seen with ketamine.

Because existing pharmacological treatments for MDD are often insufficient for many patients, the next generation of treatments needs to be more effective, rapid acting and better tolerated than currently available medications. There is extant evidence that the glutamatergic system holds considerable promise for developing the next generation of novel and mechanistically distinct agents for the treatment of MDD.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Table I
Table II
Fig. 1

Similar content being viewed by others

References

  1. Cole JO. Therapeutic efficacy of antidepressant drugs: a review. JAMA 1964; 190: 448–55

    Article  PubMed  CAS  Google Scholar 

  2. Davis JM. Efficacy of tranquilizing and antidepressant drugs. Arch Gen Psychiatry 1965; 13 (6): 552–72

    Article  PubMed  CAS  Google Scholar 

  3. Jensen K. Depressions in patients treated with reserpine for arterial hypertension. Acta Psychiatr Neurol Scand 1959; 34 (2): 195–204

    Article  PubMed  CAS  Google Scholar 

  4. Bunney Jr WE, Davis JM. Norepinephrine in depressive reactions: a review. Arch Gen Psychiatry 1965; 13 (6): 483–94

    Article  PubMed  CAS  Google Scholar 

  5. Schildkraut JJ. The catecholamine hypothesis of affective disorders: a review of supporting evidence. Am J Psychiatry 1965; 122 (5): 509–22

    PubMed  CAS  Google Scholar 

  6. Connolly KR, Thase ME. If at first you don’t succeed: a review of the evidence for antidepressant augmentation, combination and switching strategies. Drugs 2011; 71 (1): 43–64

    Article  PubMed  CAS  Google Scholar 

  7. Schechter LE, Ring RH, Beyer CE, et al. Innovative approaches for the development of antidepressant drugs: current and future strategies. Neuro Rx 2005; 2 (4): 590–611

    Article  PubMed  Google Scholar 

  8. Murray CJ, Lopez AD. Evidence-based health policy: lessons from the Global Burden of Disease Study. Science 1996; 274 (5288): 740–3

    Article  PubMed  CAS  Google Scholar 

  9. Trivedi MH, Rush AJ, Wisniewski SR, et al. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry 2006; 163 (1): 28–40

    Article  PubMed  Google Scholar 

  10. Kirsch I, Deacon BJ, Huedo-Medina TB, et al. Initial severity and antidepressant benefits: a meta-analysis of data submitted to the Food and Drug Administration. PLoS Med 2008; 5 (2): e45

  11. Horder J, Matthews P, Waldmann R. Placebo, prozac and PLoS: significant lessons for psychopharmacology. J Psy-chopharmacol 2011; 25 (10): 1277–88

    CAS  Google Scholar 

  12. Fountoulakis KN, Möller HJ. Efficacy of antidepressants: a re-analysis and re-interpretation of the Kirsch data. Int J Neuropsychopharmacol 2011; 14 (3): 405–12

    Article  PubMed  CAS  Google Scholar 

  13. Gueorguieva R, Mallinckrodt C, Krystal JH. Trajectories of depression severity in clinical trials of duloxetine: insights into antidepressant and placebo responses. Arch Gen Psychiatry 2011; 68 (12): 1227–37

    Article  PubMed  CAS  Google Scholar 

  14. Trullas R, Skolnick P. Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. Eur J Pharmacol 1990; 185 (1): 1–10

    Article  PubMed  CAS  Google Scholar 

  15. Skolnick P, Layer RT, Popik P, et al. Adaptation of N-methyl-D-aspartate (NMDA) receptors following antidepressant treatment: implications for the pharmacotherapy of depression. Pharmacopsychiatry 1996; 29 (1): 23–6

    Article  PubMed  CAS  Google Scholar 

  16. Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 2000; 47 (4): 351–4

    Article  PubMed  CAS  Google Scholar 

  17. Zarate Jr CA, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatmentresistant major depression. Arch Gen Psychiatry 2006; 63 (8): 856–64

    Article  PubMed  CAS  Google Scholar 

  18. Machado-Vieira R, Salvadore G, Luckenbaugh DA, et al. Rapid onset of antidepressant action: a new paradigm in the research and treatment of major depressive disorder. J Clin Psychiatry 2008; 69 (6): 946–58

    Article  PubMed  Google Scholar 

  19. Watkins JC, Jane DE. The glutamate story. Br J Pharmacol 2006; 147 Suppl. 1: S100–8

    PubMed  CAS  Google Scholar 

  20. Evans RH, Francis AA, Hunt K, et al. Antagonism of excitatory amino acid-induced responses and of synaptic excitation in the isolated spinal cord of the frog. Br J Pharmacol 1979; 67 (4): 591–603

    Article  PubMed  CAS  Google Scholar 

  21. Verkhratsky A, Kirchhoff F. Glutamate-mediated neuronal-glial transmission. J Anat 2007; 210 (6): 651–60

    Article  PubMed  CAS  Google Scholar 

  22. Mathew SJ, Keegan K, Smith L. Glutamate modulators as novel interventions for mood disorders. Rev Bras Psiquiatr 2005; 27 (3): 243–8

    Article  PubMed  Google Scholar 

  23. Malenka RC, Nicoll RA. Long-term potentiation: a decade of progress?. Science 1999; 285 (5435): 1870–4

    Article  PubMed  CAS  Google Scholar 

  24. Parsons CG, Danysz W, Quack G. Glutamate in CNS disorders as a target for drug development: an update. Drug News Perspect 1998; 11 (9): 523–9

    Article  PubMed  CAS  Google Scholar 

  25. Francis PT. Glutamatergic systems in Alzheimer’s disease. Int J Geriatr Psychiatry 2003; 18 Suppl. 1: S15–21

    Article  PubMed  Google Scholar 

  26. Cortese BM, Phan KL. The role of glutamate in anxiety and related disorders. CNS Spectr 2005; 10 (10): 820–30

    PubMed  Google Scholar 

  27. Fan MM, Raymond LA. N-methyl-D-aspartate (NMDA) receptor function and excitotoxicity in Huntington’s disease. Prog Neurobiol 2007; 81 (5–6): 272–93

    Article  PubMed  CAS  Google Scholar 

  28. Schoepp DD. Unveiling the functions of presynaptic metabotropic glutamate receptors in the central nervous system. J Pharmacol Exp Ther 2001; 299 (1): 12–20

    PubMed  CAS  Google Scholar 

  29. Kim JS, Schmid-Burgk W, Claus D, et al. Increased serum glutamate in depressed patients. Arch Psychiatr Nervenkr 1982; 232 (4): 299–304

    Article  PubMed  CAS  Google Scholar 

  30. Altamura CA, Mauri MC, Ferrara A, et al. Plasma and platelet excitatory amino acids in psychiatric disorders. Am J Psychiatry 1993; 150 (11): 1731–3

    PubMed  CAS  Google Scholar 

  31. Altamura CA, Mauri MC, Ferrara A, et al. Plasma and platelet amino acid concentrations in patients affected by major depression and under fluvoxamine treatment. Neuropsychobiology 1998; 37 (3): 124–9

    Article  Google Scholar 

  32. Mitani H, Shirayama Y, Yamada T, et al. Correlation between plasma levels of glutamate, alanine and serine with severity of depression. Prog Neuropsychopharmacol Biol Psychiatry 2006; 30 (6): 1155–8

    Article  PubMed  CAS  Google Scholar 

  33. Levine J, Panchalingam K, Rapoport A, et al. Increased cerebrospinal fluid glutamine levels in depressed patients. Biol Psychiatry 2000; 47 (7): 586–93

    Article  PubMed  CAS  Google Scholar 

  34. Frye MA, Tsai GE, Huggins T, et al. Low cerebrospinal fluid glutamate and glycine in refractory affective disorder [published erratum appears in Biol Psychiatry 2007; 61 (10): 1221]. Biol Psychiatry 2007; 61 (2): 162–6

    Article  PubMed  CAS  Google Scholar 

  35. Francis PT, Poynton A, Lowe SL, et al. Brain amino acid concentrations and Ca2+-dependent release in intractable depression assessed antemortem. Brain Res 1989; 494 (2): 315–24

    Article  PubMed  CAS  Google Scholar 

  36. Nowak G, Ordway GA, Paul IA. Alterations in the N-methyl-D-aspartate (NMDA) receptor complex in the frontal cortex of suicide victims. Brain Res 1995; 675 (1–2): 157–64

    Article  PubMed  CAS  Google Scholar 

  37. Holemans S, De Paermentier F, Horton RW, et al. NMDA glutamatergic receptors, labelled with [3H]MK-801, in brain samples from drug-free depressed suicides. Brain Res 1993; 616 (1–2): 138–43

    Article  PubMed  CAS  Google Scholar 

  38. Altamura C, Maes M, Dai J, et al. Plasma concentrations of excitatory amino acids, serine, glycine, taurine and histidine in major depression. Eur Neuropsychopharmacol 1995; 5 Suppl.: 71–5

    Article  PubMed  CAS  Google Scholar 

  39. Maes M, Verkerk R, Vandoolaeghe E, et al. Serum levels of excitatory amino acids, serine, glycine, histidine, threonine, taurine, alanine and arginine in treatment-resistant depression: modulation by treatment with antidepressants and prediction of clinical responsivity. Acta Psychiatr Scand 1998; 97 (4): 302–8

    Article  PubMed  CAS  Google Scholar 

  40. Frye MA, Watzl J, Banakar S, et al. Increased anterior cingulate/medial prefrontal cortical glutamate and creatine in bipolar depression. Neuropsychopharmacology 2007; 32 (12): 2490–9

    Article  PubMed  CAS  Google Scholar 

  41. Hasler G, van der Veen JW, et al. Reduced prefrontal glutamate/glutamine and gamma-aminobutyric acid levels in major depression determined using proton magnetic resonance spectroscopy. Arch Gen Psychiatry 2007; 64 (2): 193–200

    Article  PubMed  CAS  Google Scholar 

  42. Sanacora G, Gueorguieva R, Epperson CN, et al. Subtypespecific alterations of gamma-aminobutyric acid and glutamate in patients with major depression. Arch Gen Psychiatry 2004; 61 (7): 705–13

    Article  PubMed  CAS  Google Scholar 

  43. Yildiz-Yesiloglu A, Ankerst DP. Neurochemical alterations of the brain in bipolar disorder and their implications for pathophysiology: a systematic review of the in vivo proton magnetic resonance spectroscopy findings. Prog Neuropsychopharmacol Biol Psychiatry 2006; 30 (6): 969–95

    Article  PubMed  CAS  Google Scholar 

  44. Yuksel C, Ongur D. Magnetic resonance spectroscopy studies of glutamate-related abnormalities in mood disorders. Biol Psychiatry 2010; 68 (9): 785–94

    Article  PubMed  CAS  Google Scholar 

  45. Salvadore G, Zarate CA Jr. Magnetic resonance spectroscopy studies of the glutamatergic system in mood disorders: a pathway to diagnosis, novel therapeutics, and personalized medicine?. Biol Psychiatry 2010 Nov 1; 68 (9): 780–2

    Article  PubMed  Google Scholar 

  46. Gorman JM, Docherty JP. A hypothesized role for dendritic remodeling in the etiology of mood and anxiety disorders. J Neuropsychiatry Clin Neurosci 2010; 22 (3): 256–64

    Article  PubMed  Google Scholar 

  47. Holmes A, Wellman CL. Stress-induced prefrontal reorganization and executive dysfunction in rodents. Neurosci Biobehav Rev 2009; 33 (6): 773–83

    Article  PubMed  Google Scholar 

  48. McEwen BS. Stress, sex, and neural adaptation to a changing environment: mechanisms of neuronal remodeling. Ann N Y Acad Sci 2010; 1204 Suppl.: E38–59

    Article  PubMed  Google Scholar 

  49. Pittenger C, Duman RS. Stress, depression, and neuroplasticity: a convergence of mechanisms. Neuropsychopharmacology 2008; 33 (1): 88–109

    Article  PubMed  CAS  Google Scholar 

  50. Lee LJ, Lo FS, Erzurumlu RS. NMDA receptor-dependent regulation of axonal and dendritic branching. J Neurosci 2005; 25 (9): 2304–11

    Article  PubMed  CAS  Google Scholar 

  51. Bessa JM, Ferreira D, Melo I, 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, 739

    Article  PubMed  CAS  Google Scholar 

  52. Ongur D, Haddad S, Prescot AP, et al. Relationship between genetic variation in the glutaminase gene GLS1 and brain glutamine/glutamate ratio measured in vivo. Biol Psychiatry 2011; 70 (2): 169–74

    Article  PubMed  CAS  Google Scholar 

  53. Sanacora G, Treccani G, Popoli M. Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology 2012; 62 (1): 63–77

    Article  PubMed  CAS  Google Scholar 

  54. Manji HK, Quiroz JA, Sporn J, et al. Enhancing neuronal plasticity and cellular resilience to develop novel, improved therapeutics for difficult-to-treat depression. Biol Psychiatry 2003; 53 (8): 707–42

    Article  PubMed  CAS  Google Scholar 

  55. Diazgranados N, Ibrahim L, Brutsche NE, 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

    Article  PubMed  CAS  Google Scholar 

  56. Zarate Jr CA, Brutsche NE, Ibrahim L, et al. Replication of ketamine’s antidepressant efficacy in bipolar depression: a randomized controlled add-on trial. Biol Psychiatry 2012; 71 (11): 939–46

    Article  PubMed  CAS  Google Scholar 

  57. Zarate Jr CA, Singh JB, Quiroz JA, et al. A double-blind, placebo-controlled study of memantine in the treatment of major depression. Am J Psychiatry 2006; 163 (1): 153–5

    Article  PubMed  Google Scholar 

  58. Anand A, Gunn AD, Barkay G, et al. Early antidepressant effect of memantine during augmentation of lamotrigine inadequate response in bipolar depression: a doubleblind, randomized, placebo-controlled trial. Bipolar Disord 2012; 14 (1): 64–70

    Article  PubMed  CAS  Google Scholar 

  59. Lenze EJ, Skidmore ER, Begley AE, et al. 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. Epub 2011 Dec 16

  60. Mathew SJ, Murrough JW, Rot M, et al. Riluzole for relapse prevention following intravenous ketamine in treatment-resistant depression: a pilot randomized, placebo-controlled continuation trial. Int J Neuropsychopharmacol 2010; 13 (1): 71–82

    Article  PubMed  CAS  Google Scholar 

  61. Ibrahim L, Diazgranados N, Franco-Chaves J, et al. Course of improvement in depressive symptoms to a single intravenous infusion of ketamine vs. add-on riluzole: results from a four-week, double-blind, placebo-controlled study. Neuropsychopharmacology 2012 May; 37 (6): 1526–33

    CAS  Google Scholar 

  62. Preskorn SH, Baker B, Kolluri S, et al. 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

    Article  PubMed  CAS  Google Scholar 

  63. Ibrahim L, DiazGranados N, Jolkovsky L, 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. Neuropsychopharmacology 2012 May; 37 (6): 1526–33

    Article  PubMed  CAS  Google Scholar 

  64. Machado-Vieira R, Manji HK, Zarate CA. The role of the tripartite glutamatergic synapse in the pathophysiology and therapeutics of mood disorders. Neuroscientist 2009; 15 (5): 525–39

    Article  PubMed  CAS  Google Scholar 

  65. Lesch KP, Schmitt A. Antidepressants and gene expression profiling: how to SNARE novel drug targets. Pharmacogenomics J 2002; 2 (6): 346–8

    Article  PubMed  CAS  Google Scholar 

  66. Meloni D, Gambarana C, De Montis MG, et al. Dizocilpine antagonizes the effect of chronic imipramine on learned helplessness in rats. Pharmacol Biochem Behav 1993; 46 (2): 423–6

    Article  PubMed  CAS  Google Scholar 

  67. Papp M, Moryl E. Antidepressant activity of noncompetitive and competitive NMDA receptor antagonists in a chronic mild stress model of depression. Eur J Pharmacol 1994; 263 (1–2): 1–7

    Article  PubMed  CAS  Google Scholar 

  68. Layer RT, Popik P, Olds T, et al. Antidepressant-like actions of the polyamine site NMDA antagonist, eliprodil (SL-82.0715). Pharmacol Biochem Behav 1995; 52 (3): 621–7

    Article  PubMed  CAS  Google Scholar 

  69. Przegaliński E, Tatarczyńska E, Dereń-Wesolek A, et al. Antidepressant-like effects of a partial agonist at strychnine-insensitive glycine receptors and a competitive NMDA receptor antagonist. Neuropharmacology 1997; 36 (1): 31–7

    Article  PubMed  Google Scholar 

  70. Nowak G, Trullas R, Layer RT, et al. Adaptive changes in the N-methyl-D-aspartate receptor complex after chronic treatment with imipramine and 1-aminocyclopropane-carboxylic acid. J Pharmacol Exp Ther 1993; 265 (3): 1380–6

    PubMed  CAS  Google Scholar 

  71. Paul IA, Layer RT, Skolnick P, et al. Adaptation of the NMDA receptor in rat cortex following chronic electroconvulsive shock or imipramine. Eur J Pharmacol 1993; 247 (3): 305–11

    Article  PubMed  CAS  Google Scholar 

  72. Paul IA, Nowak G, Layer RT, et al. Adaptation of the N-methyl-D-aspartate receptor complex following chronic antidepressant treatments. J Pharmacol Exp Ther 1994; 269 (1): 95–102

    PubMed  CAS  Google Scholar 

  73. Chaturvedi HK, Chandra D, Bapna JS. Interaction between N-methyl-D-aspartate receptor antagonists and imipramine in shock-induced depression. Indian J Exp Biol 1999; 37 (10): 952–8

    PubMed  CAS  Google Scholar 

  74. Machado-Vieira R, Salvadore G, Ibrahim LA, et al. Targeting glutamatergic signaling for the development of novel therapeutics for mood disorders. Curr Pharm Des 2009; 15 (14): 1595–611

    Article  PubMed  CAS  Google Scholar 

  75. Sanacora G, Zarate CA, Krystal JH, et al. Targeting the glutamatergic system to develop novel, improved therapeutics for mood disorders. Nat Rev Drug Discov 2008; 7 (5): 426–37

    Article  PubMed  CAS  Google Scholar 

  76. Malinow R, Malenka RC. AMPA receptor trafficking and synaptic plasticity. Annu Rev Neurosci 2002; 25: 103–26

    Article  PubMed  CAS  Google Scholar 

  77. Zhu JJ, Qin Y, Zhao M, et al. Ras and Rap control AMPA receptor trafficking during synaptic plasticity. Cell 2002; 110 (4): 443–55

    Article  PubMed  CAS  Google Scholar 

  78. Esteban JA. AMPA receptor trafficking: a road map for synaptic plasticity. Mol Interv 2003; 3 (7): 375–85

    Article  PubMed  CAS  Google Scholar 

  79. Anggono V, Huganir RL. Regulation of AMPA receptor trafficking and synaptic plasticity. Curr Opin Neurobiol. Epub 2012 Jan 2

  80. Bleakman D, Alt A, Witkin JM. AMPA receptors in the therapeutic management of depression. CNS Neurol Disord Drug Targets 2007; 6 (2): 117–26

    Article  PubMed  CAS  Google Scholar 

  81. Chourbaji S, Vogt MA, Fumagalli F, et al. AMPA receptor subunit 1 (GluR-A) knockout mice model the glutamate hypothesis of depression. FASEB J 2008; 22 (9): 3129–34

    Article  PubMed  CAS  Google Scholar 

  82. Gibbons AS, Brooks L, Scarr E, et al. AMPA receptor expression is increased post-mortem samples of the anterior cingulate from subjects with major depressive disorder. J Affect Disord 2012 Feb; 136 (3): 1232–7

    Article  PubMed  CAS  Google Scholar 

  83. Maeng S, Zarate Jr CA, Du J, 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

    Article  PubMed  CAS  Google Scholar 

  84. Zarate Jr C, Machado-Vieira R, Henter I, et al. Glutamatergic modulators: the future of treating mood disorders?. Harv Rev Psychiatry 2010; 18 (5): 293–303

    Article  PubMed  Google Scholar 

  85. Alt A, Nisenbaum ES, Bleakman D, et al. A role for AMPA receptors in mood disorders. Biochem Pharmacol 2006; 71 (9): 1273–88

    Article  PubMed  CAS  Google Scholar 

  86. O’Neill MJ, Witkin JM. AMPA receptor potentiators: application for depression and Parkinson’s disease. Curr Drug Targets 2007; 8 (5): 603–20

    Article  PubMed  Google Scholar 

  87. Bleakman D, Lodge D. Neuropharmacology of AMPA and kainate receptors. Neuropharmacology 1998; 37 (10-11): 1187-204

    Google Scholar 

  88. Borges K, Dingledine R. AMPA receptors: molecular and functional diversity. Prog Brain Res 1998; 116: 153–70

    Article  PubMed  CAS  Google Scholar 

  89. Li X, Tizzano JP, Griffey K, et al. Antidepressant-like actions of an AMPA receptor potentiator (LY392098). Neuropharmacology 2001; 40 (8): 1028–33

    Article  PubMed  CAS  Google Scholar 

  90. Knapp RJ, Goldenberg R, Shuck C, et al. Antidepressant activity of memory-enhancing drugs in the reduction of submissive behavior model. Eur J Pharmacol 2002; 440 (1): 27–35

    Article  PubMed  CAS  Google Scholar 

  91. Lindholm JS, Autio H, Vesa L, et al. The antidepressantlike effects of glutamatergic drugs ketamine and AMPA receptor potentiator LY 451646 are preserved in bdnf/heterozygous null mice. Neuropharmacology 2012; 62 (1): 391–7

    Article  PubMed  CAS  Google Scholar 

  92. Bursi R, Erdemli G, Campbell R, et al. Translational PK-PD modelling of molecular target modulation for the AMPA receptor positive allosteric modulator Org 26576. Psychopharmacology (Berl) 2011; 218 (4): 713–24

    Article  CAS  Google Scholar 

  93. Niciu MJ, Kelmendi B, Sanacora G. Overview of glutamatergic neurotransmission in the nervous system. Pharmacol Biochem Behav 2012; 100 (4): 656–64

    Article  PubMed  CAS  Google Scholar 

  94. Szewczyk B, Palucha-Poniewiera A, Poleszak E, et al. Investigational NMDA receptor modulators for depression. Expert Opin Investig Drugs 2012; 21 (1): 91–102

    Article  PubMed  CAS  Google Scholar 

  95. Krystal JH, Mathew SJ, D’Souza DC, et al. Potential psychiatric applications of metabotropic glutamate receptor agonists and antagonists. CNS Drugs 2010; 24 (8): 669–93

    Article  PubMed  CAS  Google Scholar 

  96. Harrison NL, Simmonds MA. Quantitative studies on some antagonists of N-methyl D-aspartate in slices of rat cerebral cortex. Br J Pharmacol 1985; 84 (2): 381–91

    Article  PubMed  CAS  Google Scholar 

  97. Bolshakov KV, Gmiro VE, Tikhonov DB, et al. Determinants of trapping block of N-methyl-d-aspartate receptor channels. J Neurochem 2003; 87 (1): 56–65

    Article  PubMed  CAS  Google Scholar 

  98. Narita M, Yoshizawa K, Nomura M, et al. Role of the NMDA receptor subunit in the expression of the discriminative stimulus effect induced by ketamine. Eur J Pharmacol 2001; 423 (1): 41–6

    Article  PubMed  CAS  Google Scholar 

  99. De Vry J, Jentzsch KR. Role of the NMDA receptor NR2B subunit in the discriminative stimulus effects of ketamine. Behav Pharmacol 2003; 14 (3): 229–35

    Article  PubMed  Google Scholar 

  100. Maler JM, Esselmann H, Wiltfang J, et al. Memantine inhibits ethanol-induced NMDA receptor up-regulation in rat hippocampal neurons. Brain Res 2005; 1052 (2): 156–62

    Article  PubMed  CAS  Google Scholar 

  101. Maeng S, Zarate Jr CA. The role of glutamate in mood disorders: results from the ketamine in major depression study and the presumed cellular mechanism underlying its antidepressant effects. Curr Psychiatry Rep 2007; 9 (6): 467–74

    Article  PubMed  Google Scholar 

  102. Engin E, Treit D, Dickson CT. Anxiolytic- and antidepressant-like properties of ketamine in behavioral and neurophysiological animal models. Neuroscience 2009; 161 (2): 359–69

    Article  PubMed  CAS  Google Scholar 

  103. Garcia LS, Comim CM, Valvassori SS, et al. Chronic administration of ketamine elicits antidepressant-like effects in rats without affecting hippocampal brain-derived neurotrophic factor protein levels. Basic Clin Pharmacol Toxicol 2008; 103 (6): 502–6

    Article  PubMed  CAS  Google Scholar 

  104. Garcia LS, Comim CM, Valvassori SS, 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

    Article  PubMed  CAS  Google Scholar 

  105. Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science 2010; 329 (5994): 959–64

    Article  PubMed  CAS  Google Scholar 

  106. Yilmaz A, Schulz D, Aksoy A, et al. Prolonged effect of an anesthetic dose of ketamine on behavioral despair. Pharmacol Biochem Behav 2002; 71 (1–2): 341–4

    Article  PubMed  CAS  Google Scholar 

  107. Garcia LS, Comim CM, Valvassori SS, et al. Ketamine treatment reverses behavioral and physiological alterations induced by chronic mild stress in rats. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33 (3): 450–5

    Article  PubMed  CAS  Google Scholar 

  108. Li N, Liu RJ, Dwyer JM, 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

    Article  PubMed  CAS  Google Scholar 

  109. da Silva FC, do Carmo de Oliveira Cito M, da Silva MI, et al. Behavioral alterations and pro-oxidant effect of a single ketamine administration to mice. Brain Res Bull 2010; 83 (1–2): 9–15

    Article  PubMed  CAS  Google Scholar 

  110. Rosa AO, Lin J, Calixto JB, et al. Involvement of NMDA receptors and L-arginine-nitric oxide pathway in the antidepressant-like effects of zinc in mice. Behav Brain Res 2003; 144 (1–2): 87–93

    Article  PubMed  CAS  Google Scholar 

  111. Kos T, Popik P, Pietraszek M, et al. Effect of 5-HT3 receptor antagonist MDL 72222 on behaviors induced by ketamine in rats and mice. Eur Neuropsychopharmacol 2006; 16 (4): 297–310

    Article  PubMed  CAS  Google Scholar 

  112. Chindo BA, Adzu B, Yahaya TA, et al. Ketamineenhanced immobility in forced swim test: a possible animal model for negative symptoms of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2012; 26: 26

    Google Scholar 

  113. Moghaddam B, Adams B, Verma A, et al. 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

    PubMed  CAS  Google Scholar 

  114. Green SM, Rothrock SG, Lynch EL, et al. Intramuscular ketamine for pediatric sedation in the emergency department: safety profile in 1022 cases. Ann Emerg Med 1998; 31 (6): 688–97

    Article  PubMed  CAS  Google Scholar 

  115. Howes MC. Ketamine for paediatric sedation/analgesia in the emergency department. Emerg Med J 2004; 21 (3): 275–80

    Article  PubMed  CAS  Google Scholar 

  116. Strayer RJ, Nelson LS. Adverse events associated with ketamine for procedural sedation in adults. Am J Emerg Med 2008; 26 (9): 985–1028

    Article  PubMed  Google Scholar 

  117. Jevtovic-Todorovic V, Wozniak DF, Benshoff ND, et al. A comparative evaluation of the neurotoxic properties of ketamine and nitrous oxide. Brain Res 2001; 895 (1–2): 264–7

    Article  PubMed  CAS  Google Scholar 

  118. Valentine GW, Mason GF, Gomez R, et al. The antidepressant effect of ketamine is not associated with changes in occipital amino acid neurotransmitter content as measured by [(1)H]-MRS. Psychiatry Res 2011; 191 (2): 122–7

    Article  PubMed  CAS  Google Scholar 

  119. Bunney BG, Bunney WE. Rapid-acting antidepressant strategies: mechanisms of action. Int J Neuropsychopharmacol 2011; 7: 1–19

    Google Scholar 

  120. Rot M, Collins KA, Murrough JW, et al. Safety and efficacy of repeated-dose intravenous ketamine for treatment-resistant depression. Biol Psychiatry 2010; 67 (2): 139–45

    Article  CAS  Google Scholar 

  121. Phelps LE, Brutsche N, Moral JR, et al. Family history of alcohol dependence and initial antidepressant response to an N-methyl-D-aspartate antagonist. Biol Psychiatry 2009; 65 (2): 181–4

    Article  PubMed  CAS  Google Scholar 

  122. Price RB, Nock MK, Charney DS, et al. Effects of intravenous ketamine on explicit and implicit measures of suicidality in treatment-resistant depression. Biol Psychiatry 2009; 66 (5): 522–6

    Article  PubMed  CAS  Google Scholar 

  123. DiazGranados N, Ibrahim LA, Brutsche NE, 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

    Article  PubMed  CAS  Google Scholar 

  124. Larkin GL, Beautrais AL. A preliminary naturalistic study of low-dose ketamine for depression and suicide ideation in the emergency department. Int J Neuropsychopharmacol 2011; 14 (8): 1127–31

    Article  PubMed  CAS  Google Scholar 

  125. Mathew SJ, Shah A, Lapidus K, et al. Ketamine for treatment-resistant unipolar depression: current evidence. CNS Drugs 2012; 26 (3): 189–204

    Article  PubMed  CAS  Google Scholar 

  126. Doble A. The pharmacology and mechanism of action of riluzole. Neurology 1996; 47 (6 Suppl. 4): S233-41

    Google Scholar 

  127. Du J, Suzuki K, Wei Y, et al. The anticonvulsants lamotrigine, riluzole, and valproate differentially regulate AMPA receptor membrane localization: relationship to clinical effects in mood disorders. Neuropsychopharmacology 2007; 32 (4): 793–802

    Article  PubMed  CAS  Google Scholar 

  128. Mizuta I, Ohta M, Ohta K, et al. Riluzole stimulates nerve growth factor, brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor synthesis in cultured mouse astrocytes. Neurosci Lett 2001; 310 (2-3): 117-20

    Google Scholar 

  129. Zarate CA, Manji HK. Riluzole in psychiatry: a systematic review of the literature. Expert Opin Drug Metab Toxicol 2008; 4 (9): 1223–34

    Article  PubMed  CAS  Google Scholar 

  130. Papazisis G, et al. Neuroprotection by lamotrigine in a rat model of neonatal hypoxic-ischaemic encephalopathy. Int J Neuropsychopharmacol 2008; 11 (3): 321–9

    Article  PubMed  CAS  Google Scholar 

  131. Goodnick PJ. Bipolar depression: a review of randomised clinical trials. Expert Opin Pharmacother 2007; 8 (1): 13–21

    Article  PubMed  CAS  Google Scholar 

  132. Geddes JR, Calabrese JR, Goodwin GM. Lamotrigine for treatment of bipolar depression: independent meta-analysis and meta-regression of individual patient data from five randomised trials. Br J Psychiatry 2009; 194 (1): 4–9

    Article  PubMed  Google Scholar 

  133. Anand A, Charney DS, Oren DA, et al. Attenuation of the neuropsychiatric effects of ketamine with lamotrigine: support for hyperglutamatergic effects of N-methyl-D-aspartate receptor antagonists. Arch Gen Psychiatry 2000; 57 (3): 270–6

    Article  PubMed  CAS  Google Scholar 

  134. Wang M, Yang Y, Dong Z, et al. 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

    Article  PubMed  CAS  Google Scholar 

  135. Loftis JM, Janowsky A. The N-methyl-D-aspartate receptor subunit NR2B: localization, functional properties, regulation, and clinical implications. Pharmacol Ther 2003; 97 (1): 55–85

    Article  PubMed  CAS  Google Scholar 

  136. Gogas KR. Glutamate-based therapeutic approaches: NR2B receptor antagonists. Curr Opin Pharmacol 2006; 6 (1): 68–74

    Article  PubMed  CAS  Google Scholar 

  137. Merchant RE, Bullock MR, Carmack CA, et al. A doubleblind, placebo-controlled study of the safety, tolerability and pharmacokinetics of CP-101, 606 in patients with a mild or moderate traumatic brain injury. Ann N Y Acad Sci 1999; 890: 42–50

    Article  PubMed  CAS  Google Scholar 

  138. Faries D, Herrera J, Rayamajhi J, et al. The responsiveness of the Hamilton Depression Rating Scale. J Psychiatr Res 2000; 34 (1): 3–10

    Article  PubMed  CAS  Google Scholar 

  139. Kemp AH, Gordon E, Rush AJ, et al. Improving the prediction of treatment response in depression: integration of clinical, cognitive, psychophysiological, neuroimaging, and genetic measures. CNS Spectr 2008; 13 (12): 1066–86; quiz 1087-8

    PubMed  Google Scholar 

  140. Kornhuber J, Weller M, Schoppmeyer K, et al. Amantadine and memantine are NMDA receptor antagonists with neuroprotective properties. J Neural Transm Suppl 1994; 43: 91–104

    PubMed  CAS  Google Scholar 

  141. Bormann J. Memantine is a potent blocker of N-methyl-D-aspartate (NMDA) receptor channels. Eur J Pharmacol 1989; 166 (3): 591–2

    Article  PubMed  CAS  Google Scholar 

  142. Kornhuber J, Bormann J, Retz W, et al. Memantine displaces [3H]MK-801 at therapeutic concentrations in postmortem human frontal cortex. Eur J Pharmacol 1989; 166 (3): 589–90

    Article  PubMed  CAS  Google Scholar 

  143. Kornhuber J, Bormann J, Hübers M, et al. Effects of the 1-amino-adamantanes at the MK-801-binding site of the NMDA-receptor-gated ion channel: a human postmortem brain study. Eur J Pharmacol 1991; 206 (4): 297–300

    Article  PubMed  CAS  Google Scholar 

  144. Kornhuber J, Weller M. Psychotogenicity and N-methyl-D-aspartate receptor antagonism: implications for neuroprotective pharmacotherapy. Biol Psychiatry 1997; 41 (2): 135–44

    Article  PubMed  CAS  Google Scholar 

  145. Muller WE, Mutschler E, Riederer P. Noncompetitive NMDA receptor antagonists with fast open-channel blocking kinetics and strong voltage-dependency as potential therapeutic agents for Alzheimer’s dementia. Pharmacopsychiatry 1995; 28 (4): 113–24

    Article  PubMed  CAS  Google Scholar 

  146. Lo D, Grossberg GT. Use of memantine for the treatment of dementia. Expert Rev Neuroth er 2011; 11 (10): 1359–70

    Article  CAS  Google Scholar 

  147. Parsons CG, Danysz W, Quack G. Memantine is a clinically well tolerated N-methyl-D-aspartate (NMDA) receptor antagonist: a review of preclinical data. Neuropharmacology 1999; 38 (6): 735–67

    Article  PubMed  CAS  Google Scholar 

  148. Tsai GE. Searching for rational anti N-methyl-D-aspartate treatment for depression. Arch Gen Psychiatry, 2007; 64 (9): 1099–100; author reply 1100-1

    Article  PubMed  Google Scholar 

  149. Muhonen LH, Lönnqvist J, Juva K, et al. Double-blind, randomized comparison of memantine and escitalopram for the treatment of major depressive disorder comorbid with alcohol dependence. J Clin Psychiatry 2008; 69 (3): 392–9

    Article  PubMed  CAS  Google Scholar 

  150. Holter SM, Danysz W, Spanagel R. Evidence for alcohol anti-craving properties of memantine. Eur J Pharmacol 1996; 314 (3): R1–2

    Article  PubMed  CAS  Google Scholar 

  151. Bachteler D, Economidou D, Danysz W, et al. The effects of acamprosate and neramexane on cue-induced reinstatement of ethanol-seeking behavior in rat. Neuropsychopharmacology 2005; 30 (6): 1104–10

    Article  PubMed  CAS  Google Scholar 

  152. Piasecki J, Koros E, Dyr W, et al. Ethanol-reinforced behaviour in the rat: effects of uncompetitive NMDA receptor antagonist, memantine. Eur J Pharmacol 1998; 354 (2–3): 135–43

    Article  PubMed  CAS  Google Scholar 

  153. Escher T, Call SB, Blaha CD, et al. Behavioral effects of aminoadamantane class NMDA receptor antagonists on schedule-induced alcohol and self-administration of water in mice. Psychopharmacology (Berl) 2006; 187 (4): 424–34

    Article  CAS  Google Scholar 

  154. Boden JM, Fergusson DM. Alcohol and depression. Addiction 2011; 106 (5): 906–14

    Article  PubMed  Google Scholar 

  155. Conner KR, Pinquart M, Gamble SA. Meta-analysis of depression and substance use among individuals with alcohol use disorders. J Subst Abuse Treat 2009; 37 (2): 127–37

    Article  PubMed  Google Scholar 

  156. Nagy J. Renaissance of NMDA receptor antagonists: do they have a role in the pharmacotherapy for alcoholism?. IDrugs 2004; 7 (4): 339–50

    PubMed  CAS  Google Scholar 

  157. Petrakis IL, Limoncelli D, Gueorguieve R, et al. Altered NMDA glutamate receptor antagonist response in individuals with a family vulnerability to alcoholism. Am J Psychiatry 2004; 161 (10): 1776–82

    Article  PubMed  Google Scholar 

  158. Bäckström P, Bachteler D, Koch S, et al. mGluR5 antagonist MPEP reduces ethanol-seeking and relapse behavior. Neuropsychopharmacology 2004; 29 (5): 921–8

    Article  PubMed  CAS  Google Scholar 

  159. Krupitsky EM, Rudenko AA, Burakov AM, et al. Antiglutamatergic strategies for ethanol detoxification: comparison with placebo and diazepam. Alcohol Clin Exp Res 2007; 31 (4): 604–11

    PubMed  CAS  Google Scholar 

  160. Rogóz Z, Skuza G, Maj J, et al. Synergistic effect of uncompetitive NMDA receptor antagonists and antidepressant drugs in the forced swimming test in rats. Neuropharmacology 2002; 42 (8): 1024–30

    Article  PubMed  Google Scholar 

  161. Huber TJ, Dietrich DE, Emrich HM. Possible use of amantadine in depression. Pharmacopsychiatry 1999; 32 (2): 47–55

    Article  PubMed  CAS  Google Scholar 

  162. Rogóz Z, Kubera M, Rogóz K, et al. Effect of coadministration of fluoxetine and amantadine on immunoendocrine parameters in rats subjected to a forced swimming test. Pharmacol Rep 2009; 61 (6): 1050–60

    PubMed  Google Scholar 

  163. Kubera M, Basta-Kairn A, Budziszewska B, et al. Effect of amantadine and imipramine on immunological parameters of rats subjected to a forced swimming test. Int J Neuropsychopharmacol 2006; 9 (3): 297–305

    Article  PubMed  CAS  Google Scholar 

  164. Maj J, Rogoz Z. Synergistic effect of amantadine and imipramine in the forced swimming test. Pol J Pharmacol 2000; 52 (2): 111–4

    PubMed  CAS  Google Scholar 

  165. Dietrich DE, Bode L, Spannhuth CW, et al. Amantadine in depressive patients with Borna disease virus (BDV) infection: an open trial. Bipolar Disord 2000; 2 (1): 65–70

    Article  PubMed  CAS  Google Scholar 

  166. Ferszt R, Kühl KP, Bode L, et al. Amantadine revisited: an open trial of amantadinesulfate treatment in chronically depressed patients with Borna disease virus infection. Pharmacopsychiatry 1999; 32 (4): 142–7

    Article  PubMed  CAS  Google Scholar 

  167. Stryjer R, Strous RD, Shaked G, et al. Amantadine as augmentation therapy in the management of treatmentresistant depression. Int Clin Psychopharmacol 2003; 18 (2): 93–6

    Article  PubMed  Google Scholar 

  168. Rogóz Z, Skuza G, Daniel WA, et al. Amantadine as an additive treatment in patients suffering from drugresistant unipolar depression. Pharmacol Rep 2007; 59 (6): 778–84

    PubMed  Google Scholar 

  169. Autry AE, Adachi M, Nosyreva E, et al. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature 2011; 475 (7354): 91–5

    Article  PubMed  CAS  Google Scholar 

  170. Hoeffer CA, Klann E. mTOR signaling: at the crossroads of plasticity, memory and disease. Trends Neurosci 2010; 33 (2): 67–75

    Article  PubMed  CAS  Google Scholar 

  171. Duman RS, Li N, Liu RJ, et al. Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology 2012; 62 (1): 35–41

    Article  PubMed  CAS  Google Scholar 

  172. Cryan JF, O’eary OF. Neuroscience: a glutamate pathway to faster-acting antidepressants?. Science 2010; 329 (5994): 913–4

    Article  PubMed  CAS  Google Scholar 

  173. Ravikumar B, Rubinsztein DC. Can autophagy protect against neurodegeneration caused by aggregate-prone proteins?. Neuroreport 2004; 15 (16): 2443–5

    Article  PubMed  Google Scholar 

  174. Pickford F, Masliah E, Brtischgi M, et al. The autophagyrelated protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J Clin Invest 2008; 118 (6): 2190–9

    PubMed  CAS  Google Scholar 

  175. 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

    Article  PubMed  CAS  Google Scholar 

  176. Kaidanovich-Beilin O, Milman A, Weizman A, et al. Rapid antidepressive-like activity of specific glycogen synthase kinase-3 inhibitor and its effect on beta-catenin in mouse hippocampus. Biol Psychiatry 2004; 55 (8): 781–4

    Article  PubMed  CAS  Google Scholar 

  177. O’Brien WT, Harper AD, Jové F, et al. Glycogen synthase kinase-3beta haploinsufficiency mimics the behavioral and molecular effects of lithium. J Neurosci 2004; 24 (30): 6791–8

    Article  PubMed  CAS  Google Scholar 

  178. Beaulieu JM, Sotnikova TD, Yao WD, et al. Lithium antagonizes dopamine-dependent behaviors mediated by an AKT/glycogen synthase kinase 3 signaling cascade. Proc Natl Acad Sci USA. 2004; 101 (14): 5099–104

    Article  PubMed  CAS  Google Scholar 

  179. Gould TD, Chen G, Manji HK. In vivo evidence in the brain for lithium inhibition of glycogen synthase kinase-3. Neuropsychopharmacology 2004; 29 (1): 32–8

    Article  PubMed  CAS  Google Scholar 

  180. Polter A, Beurel E, Yang S, et al. Deficiency in the inhibitory serine-phosphorylation of glycogen synthase kinase-3 increases sensitivity to mood disturbances. Neuropsychopharmacology 2010; 35 (8): 1761–74

    PubMed  CAS  Google Scholar 

  181. Jope RS, Johnson GV. The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci 2004; 29 (2): 95–102

    Article  PubMed  CAS  Google Scholar 

  182. Rayasam GV, Tulasi VK, Sodhi R, et al. Glycogen synthase kinase 3: more than a namesake. Br J Pharmacol 2009; 156 (6): 885–98

    Article  PubMed  CAS  Google Scholar 

  183. Shimizu E, Hashimoto K, Iyo M. Ethnic difference of the BDNF 196G/A (val66met) polymorphism frequencies: the possibility to explain ethnic mental traits. Am J Med Genet B Neuropsychiatr Genet 2004; 126B (1): 122-3

    Google Scholar 

  184. Casey BJ, Glatt CE, Tottenham N, et al. Brain-derived neurotrophic factor as a model system for examining gene by environment interactions across development. Neuroscience 2009; 164 (1): 108–20

    Article  PubMed  CAS  Google Scholar 

  185. Egan MF, Kojima M, Callicott JH, et al. The BDNF val66met polymorphism affects activity-dependent secretion of BDNF and human memory and hippocampal function. Cell 2003; 112 (2): 257–69

    Article  PubMed  CAS  Google Scholar 

  186. Neves-Pereira M, Mundo E, Muglia P, et al. The brainderived neurotrophic factor gene confers susceptibility to bipolar disorder: evidence from a family-based association study. Am J Hum Genet 2002; 71 (3): 651–5

    Article  PubMed  CAS  Google Scholar 

  187. Sklar P, Gabriel SB, McInnis MG, et al. Family-based association study of 76 candidate genes in bipolar disorder: BDNF is a potential risk locus. Brain-derived neutrophic factor. Mol Psychiatry 2002; 7 (6): 579–93

    Article  CAS  Google Scholar 

  188. Sen S, Nesse RM, Stoltenberg SF, et al. A BDNF coding variant is associated with the NEO personality inventory domain neuroticism, a risk factor for depression. Neuropsychopharmacology 2003; 28 (2): 397–401

    Article  PubMed  CAS  Google Scholar 

  189. Ribasés M, Gratacòs M, Fernández-Aranda F, et al. Association of BDNF with anorexia, bulimia and age of onset of weight loss in six European populations. Hum Mol Genet 2004; 13 (12): 1205–12

    Article  PubMed  Google Scholar 

  190. Sun M, Liu L, Yang Y, et al. Association study of brainderived neurotrophic factor Val66Met polymorphism and clinical characteristics of first episode schizophrenia [in Chinese]. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 2012; 29 (2): 155–8

    PubMed  CAS  Google Scholar 

  191. Autry AE, Monteggia LM. Brain-derived neurotrophic factor and neuropsychiatric disorders. Pharmacol Rev 2012; 64 (2): 238–58

    Article  PubMed  CAS  Google Scholar 

  192. Machado-Vieira R, Yuan P, Brutsche N, et al. Brainderived neurotrophic factor and initial antidepressant response to an N-methyl-D-aspartate antagonist. J Clin Psychiatry 2009; 70 (12): 1662–6

    Article  PubMed  CAS  Google Scholar 

  193. Ricci V, Martinotti G, Gelfo F, et al. Chronic ketamine use increases serum levels of brain-derived neurotrophic factor. Psychopharmacology (Berl) 2011; 215 (1): 143–8

    Article  CAS  Google Scholar 

  194. Inamura N, Nawa H, Takei N. Enhancement of translation elongation in neurons by brain-derived neurotrophic factor: implications for mammalian target of rapamycin signaling. J Neurochem 2005; 95 (5): 1438–45

    Article  PubMed  CAS  Google Scholar 

  195. Slipczuk L, Bekinschtein P, Katche C, et al. BDNF activates mTOR to regulate GluR1 expression required for memory formation. PLoS ONE 2009; 4 (6): e6007

    Article  PubMed  CAS  Google Scholar 

  196. Takei N, Inamura N, Kawamura M, et al. Brain-derived neurotrophic factor induces mammalian target of rapamy-cin-dependent local activation of translation machinery and protein synthesis in neuronal dendrites. J Neurosci 2004; 24 (44): 9760–9

    Article  PubMed  CAS  Google Scholar 

  197. Laje G, Lally N, Mathews D, et al. Brain-derived neurotrophic factor Val66Met polymorphism and antidepressant efficacy of ketamine in depressed patients. Biol Psychiatry. In press

  198. Liu RJ, Lee FS, Li XY, et al. Brain-derived neurotrophic factor Val66Met allele impairs basal and ketaminestimulated synaptogenesis in prefrontal cortex. Biol Psychiatry 2012; 71 (11): 996–1005

    Article  PubMed  CAS  Google Scholar 

  199. Murrough JW. Ketamine as a novel antidepressant: from synapse to behavior. Clin Pharmacol Ther 2012; 91 (2): 303–9

    Article  PubMed  CAS  Google Scholar 

  200. Cornwell BR, Salvadore G, Furey M, et al. Synaptic potentiation is critical for rapid antidepressant response to ketamine in treatment-resistant major depression. Biol Psychiatry. Epub 2012 Apr 20

  201. Li X, Witkin JM, Need AB, et al. Enhancement of antidepressant potency by a potentiator of AMPA receptors. Cell Mol Neurobiol 2003; 23 (3): 419–30

    Article  PubMed  CAS  Google Scholar 

  202. Maes M, Bosmans E, Suy E, et al. Immune disturbances during major depression: upregulated expression of interleukin-2 receptors. Neuropsychobiology 1990; 24 (3): 115–20

    Article  PubMed  Google Scholar 

  203. Maes M, Bosmans E, Suy E, et al. Depression-related disturbances in mitogen-induced lymphocyte responses and interleukin-1 beta and soluble interleukin-2 receptor production. Acta Psychiatr Scand 1991; 84 (4): 379–86

    Article  PubMed  CAS  Google Scholar 

  204. Maes M, Lambrechts J, Bosmans E, et al. Evidence for a systemic immune activation during depression: results of leukocyte enumeration by flow cytometry in conjunction with monoclonal antibody staining. Psychol Med 1992; 22 (1): 45–53

    Article  PubMed  CAS  Google Scholar 

  205. Bufalino C, Hepgul N, Aguglia E, et al. The role of immune genes in the association between depression and inflammation: a review of recent clinical studies. Brain Behav Immun. Epub 2012 May 8

  206. Eyre H, Baune BT. Neuroplastic changes in depression: a role for the immune system. Psychoneuroendocrinology. Epub 2012 Apr 21

  207. Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiatry 2006; 59 (12): 1116–27

    Article  PubMed  CAS  Google Scholar 

  208. Catena-Dell’Osso M, Bellantuono C, Consoli G, et al. Inflammatory and neurodegenerative pathways in depression: a new avenue for antidepressant development?. Curr Med Chem 2011; 18 (2): 245–55

    Article  PubMed  Google Scholar 

  209. Gardner A, Boles RG. Beyond the serotonin hypothesis: mitochondria, inflammation and neurodegeneration in major depression and affective spectrum disorders. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35 (3): 730–43

    Article  PubMed  CAS  Google Scholar 

  210. Maes M, et al. The inflammatory & neurodegenerative (I&ND) hypothesis of depression: leads for future research and new drug developments in depression. Metab Brain Dis 2009; 24 (1): 27–53

    Article  PubMed  CAS  Google Scholar 

  211. Leonard B, Maes M. Mechanistic explanations how cellmediated immune activation, inflammation and oxidative and nitrosative stress pathways and their sequels and concomitants play a role in the pathophysiology of unipolar depression. Neurosci Biobehav Rev 2012; 36 (2): 764–85

    Article  PubMed  CAS  Google Scholar 

  212. Moir AT, Eccleston D. The effects of precursor loading in the cerebral metabolism of 5-hydroxyindoles. J Neurochem 1968; 15 (10): 1093–108

    Article  PubMed  CAS  Google Scholar 

  213. Maes M, Leonard BE, Myint AM, et al. The new ‘5-HT’ hypothesis of depression: cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35 (3): 702–21

    Article  PubMed  CAS  Google Scholar 

  214. Muller N, Schwarz MJ. The immune-mediated alteration of serotonin and glutamate: towards an integrated view of depression. Mol Psychiatry 2007; 12 (11): 988–1000

    Article  PubMed  CAS  Google Scholar 

  215. Moylan S, Maes M, Wray NR, et al. The neuroprogressive nature of major depressive disorder: pathways to disease evolution and resistance, and therapeutic implications. Mol Psychiatry. Epub 2012 Apr 24

  216. Koga K, Ogata M, Takenaka I, et al. Ketamine suppresses tumor necrosis factor-alpha activity and mortality in carrageenan-sensitized endotoxin shock model. Circ Shock 1994; 44 (3): 160–8

    PubMed  CAS  Google Scholar 

  217. Taniguchi T, Shibata K, Yamamoto K. Ketamine inhibits endotoxin-induced shock in rats. Anesthesiology 2001; 95 (4): 928–32

    Article  PubMed  CAS  Google Scholar 

  218. Taniguchi T, Takemoto Y, Kanakura H, et al. The doserelated effects of ketamine on mortality and cytokine responses to endotoxin-induced shock in rats. Anesth Analg 2003; 97 (6): 1769–72

    Article  PubMed  CAS  Google Scholar 

  219. Suliburk JW, Helmer KS, Gonzalez EA, et al. Ketamine attenuates liver injury attributed to endotoxemia: role of cyclooxygenase-2. Surgery 2005; 138 (2): 134–40

    Article  PubMed  Google Scholar 

  220. Welters ID, Hafer G, Menzebach A, et al. Ketamine inhibits transcription factors activator protein 1 and nuclear factor-kappaB, interleukin-8 production, as well as CD11b and CD16 expression: studies in human leukocytes and leukocytic cell lines. Anesth Analg 2010; 110 (3): 934–41

    Article  PubMed  CAS  Google Scholar 

  221. Albensi BC, Mattson MP. Evidence for the involvement of TNF and NF-kappaB in hippocampal synaptic plasticity. Synapse 2000; 35 (2): 151–9

    Article  PubMed  CAS  Google Scholar 

  222. Gutierrez H, Davies AM. Regulation of neural process growth, elaboration and structural plasticity by NF-kappaB. Trends Neurosci 2011; 34 (6): 316–25

    Article  PubMed  CAS  Google Scholar 

  223. Vallabhapurapu S, Karin M. Regulation and function of NF-kappaB transcription factors in the immune system. Annu Rev Immunol 2009; 27: 693–733

    Article  PubMed  CAS  Google Scholar 

  224. Roytblat L, Talmor D, Rachinsky M, et al. Ketamine attenuates the interleukin-6 response after cardiopulmonary bypass. Anesth Analg 1998; 87 (2): 266–71

    PubMed  CAS  Google Scholar 

  225. Bartoc C, Frumento RJ, Jalbout M, et al. A randomized, double-blind, placebo-controlled study assessing the antiinflammatory effects of ketamine in cardiac surgical patients. J Cardiothorac Vasc Anesth 2006; 20 (2): 217–22

    Article  PubMed  CAS  Google Scholar 

  226. Cho JE, Shim JK, Choi YS, et al. Effect of low-dose ketamine on inflammatory response in off-pump coronary artery bypass graft surgery. Br J Anaesth 2009; 102 (1): 23–8

    Article  PubMed  CAS  Google Scholar 

  227. Carlson PJ, Singh JB, Zarate Jr CA, et al. Neural circuitry and neuroplasticity in mood disorders: insights for novel therapeutic targets. NeuroRx 2006; 3 (1): 22–41

    Article  PubMed  CAS  Google Scholar 

  228. Javitt DC, Schoepp D, Kalivas PW, et al. Translating glutamate: from pathophysiology to treatment. Sci Transl Med 2011; 3 (102): 102mr2

    Article  Google Scholar 

  229. Orloff J, Douglas F, Pinheiro J, et al. The future of drug development: advancing clinical trial design. Nat Rev Drug Discov 2009; 8 (12): 949–57

    PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors gratefully acknowledge the support of the Intramural Research Program of the National Institute of Mental Health, National Institutes of Health (IRP-NIMH-NIH; Bethesda, MD, USA), and thank the 7SE Research Unit of the NIMH-NIH for their support.

Role of funding source: This review was supported by the IRP-NIMH-NIH. The NIMH had no further role in the writing of this review, or in the decision to submit the paper for publication.

Financial disclosures: The authors gratefully acknowledge the support of the IRP-NIMH-NIH, and the NARSAD Independent Investigator Award and Brain and Behavior Foundation Bipolar Research Award (Dr Zarate). Dr Mathews and Ioline Henter have no conflict of interest to disclose, financial or otherwise. Dr Zarate is listed as a co-inventor on a patent application for the use of ketamine and its metabolites in major depression. Dr Zarate has assigned his rights in the patent to the US Government but will share a percentage of any royalties that may be received by the Government.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carlos A. Zarate Jr.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Mathews, D.C., Henter, I.D. & Zarate, C.A. Targeting the Glutamatergic System to Treat Major Depressive Disorder. Drugs 72, 1313–1333 (2012). https://doi.org/10.2165/11633130-000000000-00000

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/11633130-000000000-00000

Keywords

Navigation