Altered γ-aminobutyric acid neurotransmission in major depressive disorder: a critical review of the supporting evidence and the influence of serotonergic antidepressants

Alan L Pehrson, Connie Sanchez, Alan L Pehrson, Connie Sanchez

Abstract

Evidence suggesting that central nervous system γ-aminobutyric acid (GABA) concentrations are reduced in patients with major depressive disorder (MDD) has been present since at least 1980, and this idea has recently gained support from more recent magnetic resonance spectroscopy data. These observations have led to the assumption that MDD's underlying etiology is tied to an overall reduction in GABA-mediated inhibitory neurotransmission. In this paper, we review the mechanisms that govern GABA and glutamate concentrations in the brain, and provide a comprehensive and critical evaluation of the clinical data supporting reduced GABA neurotransmission in MDD. This review includes an evaluation of magnetic resonance spectroscopy data, as well as data on the expression and function of the GABA-synthesizing enzyme glutamic acid decarboxylase, GABA neuron-specific cell markers, such as parvalbumin, calretinin and calbindin, and the GABAA and GABAB receptors in clinical MDD populations. We explore a potential role for glial pathology in MDD-related reductions in GABA concentrations, and evidence of a connection between neurosteroids, GABA neurotransmission, and hormone-related mood disorders. Additionally, we investigate the effects of GABAergic pharmacological agents on mood, and demonstrate that these compounds have complex effects that do not universally support the idea that reduced GABA neurotransmission is at the root of MDD. Finally, we discuss the connections between serotonergic and GABAergic neurotransmission, and show that two serotonin-focused antidepressants - the selective serotonin-reuptake inhibitor fluoxetine and the multimodal antidepressant vortioxetine - modulate GABA neurotransmission in opposing ways, despite both being effective MDD treatments. Altogether, this review demonstrates that there are large gaps in our understanding of the relationship between GABA physiology and MDD, which must be remedied with more data from well-controlled empirical studies. In conclusion, this review suggests that the simplistic notion that MDD is caused by reduced GABA neurotransmission must be discarded in favor of a more nuanced and complex model of the role of inhibitory neurotransmission in MDD.

Keywords: 5-HT; GABA; major depressive disorder; serotonin.

Figures

Figure 1
Figure 1
Amino acid neurotransmitter synthesis and catabolism. The synthesis and catabolism of GABA and glutamate are tightly interconnected in the brain. Abbreviations: αKGDH, α-ketoglutarate dehydrogenase; AAT, aspartate aminotransferase; CoA, coenzyme A; Cys, cysteine; GABA, γ-aminobutyric acid; GDH, glutamate dehydrogenase; GABA-T, GABA transaminase; GAD, glutamic acid dehydrogenase; GCL, γ-glutamyl cysteine ligase; Gln, glutamine; Glu, glutamyl; Gly, glycine; GSH, glutathione; GHB, γ-hydroxybutyric acid; GGT, γ-glutamyl transferase; GGCT, γ-glutamyl cyclotransferase; OPLAH, 5-oxoprolinase (adenosine triphosphate-hydrolyzing); SSADH, succinic semialdehyde dehydrogenase; SSAR, succinic semialdehyde reductase.
Figure 2
Figure 2
Serotonergic influence on GABAergic neurotransmission in limbic system brain regions. Serotonergic heteroreceptors expressed on GABAergic interneurons and glutamatergic principal cells can modulate the excitatory state of neural networks associated with the control of cognitive function and mood. Vortioxetine may be an example of a drug that inhibits GABA neurotransmission via serotonergic mechanisms. Question marks denote receptors where expression on GABAergic interneurons has been indirectly suggested but no immunohistochemical verification exists for limbic brain regions. Abbreviations: ag, agonist; antag, antagonist; GABA, γ-aminobutyric acid; GLU, glutamate; 5-HT, 5-hydroxytryptamine; Vor, vortioxetine.

References

    1. World Health Organization Depression. 2012. [Accessed September 11, 2014]. Available from: .
    1. McIntyre RS, Cha DS, Soczynska JK, et al. Cognitive deficits and functional outcomes in major depressive disorder: determinants, substrates, and treatment interventions. Depress Anxiety. 2013;30(6):515–527.
    1. Birnbaum HG, Kessler RC, Kelley D, Ben-Hamadi R, Joish VN, Greenberg PE. Employer burden of mild, moderate, and severe major depressive disorder: mental health services utilization and costs, and work performance. Depress Anxiety. 2010;27(1):78–89.
    1. Jaeger J, Berns S, Uzelac S, Davis-Conway S. Neurocognitive deficits and disability in major depressive disorder. Psychiatry Res. 2006;145(1):39–48.
    1. Charney DS. Monoamine dysfunction and the pathophysiology and treatment of depression. J Clin Psychiatry. 1998;59(Suppl 14):11–14.
    1. Carotenuto M, Esposito M, Parisi L, et al. Depressive symptoms and childhood sleep apnea syndrome. Neuropsychiatric Dis Treat. 2012;8:369–373.
    1. Zarate C, Duman RS, Liu G, Sartori S, Quiroz J, Murck H. New paradigms for treatment-resistant depression. Ann N Y Acad Sci. 2013;1292:21–31.
    1. Martin DL, Rimvall K. Regulation of gamma-aminobutyric acid synthesis in the brain. J Neurochem. 1993;60(2):395–407.
    1. Soghomonian JJ, Martin DL. Two isoforms of glutamate decarboxylase: why? Trends Pharmacol Sci. 1998;19(12):500–505.
    1. Pinal CS, Tobin AJ. Uniqueness and redundancy in GABA production. Perspect Dev Neurobiol. 1998;5(2–3):109–118.
    1. Martin DL, Barke KE. Are GAD65 and GAD67 associated with specific pools of GABA in brain? Perspect Dev Neurobiol. 1998;5(2–3):119–129.
    1. Rowley NM, Madsen KK, Schousboe A, White HS. Glutamate and GABA synthesis, release, transport and metabolism as targets for seizure control. Neurochem Int. 2012;61(4):546–558.
    1. Koga M, Serritella AV, Messmer MM, et al. Glutathione is a physiologic reservoir of neuronal glutamate. Biochem Biophys Res Commun. 2011;409(4):596–602.
    1. Behar KL, Rothman DL. In vivo nuclear magnetic resonance studies of glutamate-gamma-aminobutyric acid-glutamine cycling in rodent and human cortex: the central role of glutamine. J Nutr. 2001;131(9 Suppl):2498S–2504S. discussion 2523S–2524S.
    1. Rae C, Hare N, Bubb WA, et al. Inhibition of glutamine transport depletes glutamate and GABA neurotransmitter pools: further evidence for metabolic compartmentation. J Neurochem. 2003;85(2):503–514.
    1. Gold BI, Bowers MB, Jr, Roth RH, Sweeney DW. GABA levels in CSF of patients with psychiatric disorders. Am J Psychiatry. 1980;137(3):362–364.
    1. Kugaya A, Sanacora G, Verhoeff NP, et al. Cerebral benzodiazepine receptors in depressed patients measured with [123I]iomazenil SPECT. Biol Psychiatry. 2003;54(8):792–799.
    1. Price RB, Shungu DC, Mao X, et al. Amino acid neurotransmitters assessed by proton magnetic resonance spectroscopy: relationship to treatment resistance in major depressive disorder. Biol Psychiatry. 2009;65(9):792–800.
    1. Sanacora G, Gueorguieva R, Epperson CN, et al. Subtype-specific alterations of gamma-aminobutyric acid and glutamate in patients with major depression. Arch Gen Psychiatry. 2004;61(7):705–713.
    1. Sanacora G, Mason GF, Rothman DL, et al. Reduced cortical gamma-aminobutyric acid levels in depressed patients determined by proton magnetic resonance spectroscopy. Arch Gen Psychiatry. 1999;56(11):1043–1047.
    1. Hasler G, van der Veen JW, Tumonis T, Meyers N, Shen J, Drevets WC. 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.
    1. 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):663–665.
    1. Sanacora G, Mason GF, Rothman DL, et al. Increased cortical GABA concentrations in depressed patients receiving ECT. Am J Psychiatry. 2003;160(3):577–579.
    1. Hasler G, Neumeister A, van der Veen JW, et al. Normal prefrontal gamma-aminobutyric acid levels in remitted depressed subjects determined by proton magnetic resonance spectroscopy. Biol Psychiatry. 2005;58(12):969–973.
    1. Shaw A, Brealy J, Richardson H, et al. Marked reductions in visual evoked responses but not gamma-aminobutyric acid concentrations or gamma-band measures in remitted depression. Biol Psychiatry. 2013;73(7):691–698.
    1. Sanacora G, Fenton LR, Fasula MK, et al. Cortical gamma-aminobutyric acid concentrations in depressed patients receiving cognitive behavioral therapy. Biol Psychiatry. 2006;59(3):284–286.
    1. Bhagwagar Z, Wylezinska M, Jezzard P, et al. Reduction in occipital cortex gamma-aminobutyric acid concentrations in medication-free recovered unipolar depressed and bipolar subjects. Biol Psychiatry. 2007;61(6):806–812.
    1. Bhagwagar Z, Wylezinska M, Jezzard P, et al. Low GABA concentrations in occipital cortex and anterior cingulate cortex in medication-free, recovered depressed patients. Int J Neuropsychopharmacol. 2008;11(2):255–260.
    1. Herrera-Guzmán I, Gudayol-Ferré E, Herrera-Guzmán D, Guàrdia-Olmos J, Hinojosa-Calvo E, Herrera-Abarca JE. Effects of selective serotonin reuptake and dual serotonergic-noradrenergic reuptake treatments on memory and mental processing speed in patients with major depressive disorder. J Psychiatr Res. 2009;43(9):855–863.
    1. Karolewicz B, Maciag D, O’Dwyer G, Stockmeier CA, Feyissa AM, Rajkowska G. Reduced level of glutamic acid decarboxylase-67 kDa in the prefrontal cortex in major depression. Int J Neuropsychopharmacol. 2010;13(4):411–420.
    1. Fatemi SH, Stary JM, Earle JA, Araghi-Niknam M, Eagan E. GABAergic dysfunction in schizophrenia and mood disorders as reflected by decreased levels of glutamic acid decarboxylase 65 and 67 kDa and Reelin proteins in cerebellum. Schizophr Res. 2005;72(2–3):109–122.
    1. Guidotti A, Auta J, Davis JM, et al. Decrease in reelin and glutamic acid decarboxylase67 (GAD67) expression in schizophrenia and bipolar disorder: a postmortem brain study. Arch Gen Psychiatry. 2000;57(11):1061–1069.
    1. Gilabert-Juan J, Varea E, Guirado R, Blasco-Ibáñez JM, Crespo C, Nácher J. Alterations in the expression of PSA-NCAM and synaptic proteins in the dorsolateral prefrontal cortex of psychiatric disorder patients. Neurosci Lett. 2012;530(1):97–102.
    1. Sibille E, Morris HM, Kota RS, Lewis DA. GABA-related transcripts in the dorsolateral prefrontal cortex in mood disorders. Int J Neuropsychopharmacol. 2011;14(6):721–734.
    1. Bielau H, Steiner J, Mawrin C, et al. Dysregulation of GABAergic neurotransmission in mood disorders: a postmortem study. Ann N Y Acad Sci. 2007;1096:157–169.
    1. Gos T, Günther K, Bielau H, et al. Suicide and depression in the quantitative analysis of glutamic acid decarboxylase-immunoreactive neuropil. J Affect Disord. 2009;113(1–2):45–55.
    1. Gos T, Steiner J, Bielau H, et al. Differences between unipolar and bipolar I depression in the quantitative analysis of glutamic acid decarboxylase-immunoreactive neuropil. Eur Arch Psychiatry Clin Neurosci. 2012;262(8):647–655.
    1. Cheetham SC, Crompton MR, Katona CL, Parker SJ, Horton RW. Brain GABAA/benzodiazepine binding sites and glutamic acid decarboxylase activity in depressed suicide victims. Brain Res. 1988;460(1):114–123.
    1. Lund JS, Lewis DA. Local circuit neurons of developing and mature macaque prefrontal cortex: Golgi and immunocytochemical characteristics. J Comp Neurol. 1993;328(2):282–312.
    1. Rajkowska G, O’Dwyer G, Teleki Z, Stockmeier CA, Miguel-Hidalgo JJ. GABAergic neurons immunoreactive for calcium binding proteins are reduced in the prefrontal cortex in major depression. Neuropsychopharmacology. 2007;32(2):471–482.
    1. Maciag D, Hughes J, O’Dwyer G, et al. Reduced density of calbindin immunoreactive GABAergic neurons in the occipital cortex in major depression: relevance to neuroimaging studies. Biol Psychiatry. 2010;67(5):465–470.
    1. Beasley CL, Zhang ZJ, Patten I, Reynolds GP. Selective deficits in prefrontal cortical GABAergic neurons in schizophrenia defined by the presence of calcium-binding proteins. Biol Psychiatry. 2002;52(7):708–715.
    1. Reynolds GP, Beasley CL, Zhang ZJ. Understanding the neurotransmitter pathology of schizophrenia: selective deficits of subtypes of cortical GABAergic neurons. J Neural Transm. 2002;109(5–6):881–889.
    1. Guilloux JP, Douillard-Guilloux G, Kota R, et al. Molecular evidence for BDNF- and GABA-related dysfunctions in the amygdala of female subjects with major depression. Mol Psychiatry. 2012;17(11):1130–1142.
    1. Tripp A, Kota RS, Lewis DA, Sibille E. Reduced somatostatin in subgenual anterior cingulate cortex in major depression. Neurobiol Dis. 2011;42(1):116–124.
    1. Rajkowska G, Miguel-Hidalgo JJ, Wei J, et al. Morphometric evidence for neuronal and glial prefrontal cell pathology in major depression. Biol Psychiatry. 1999;45(9):1085–1098.
    1. Rajkowska G, Stockmeier CA. Astrocyte pathology in major depressive disorder: insights from human postmortem brain tissue. Curr Drug Targets. 2013;14(11):1225–1236.
    1. Sanacora G, Banasr M. From pathophysiology to novel antidepressant drugs: glial contributions to the pathology and treatment of mood disorders. Biol Psychiatry. 2013;73(12):1172–1179.
    1. Paulsen RE, Fonnum F. Role of glial cells for the basal and Ca2+-dependent K+-evoked release of transmitter amino acids investigated by microdialysis. J Neurochem. 1989;52(6):1823–1829.
    1. Yüksel C, Öngür D. Magnetic resonance spectroscopy studies of glutamate-related abnormalities in mood disorders. Biol Psychiatry. 2010;68(9):785–794.
    1. Liang SL, Carlson GC, Coulter DA. Dynamic regulation of synaptic GABA release by the glutamate-glutamine cycle in hippocampal area CA1. J Neurosci. 2006;26(33):8537–8548.
    1. Levinson AJ, Fitzgerald PB, Favalli G, Blumberger DM, Daigle M, Daskalakis ZJ. Evidence of cortical inhibitory deficits in major depressive disorder. Biol Psychiatry. 2010;67(5):458–464.
    1. Paulus W, Classen J, Cohen LG, et al. State of the art: pharmacologic effects on cortical excitability measures tested by transcranial magnetic stimulation. Brain Stimul. 2008;1(3):151–163.
    1. Choudary PV, Molnar M, Evans SJ, et al. Altered cortical glutamatergic and GABAergic signal transmission with glial involvement in depression. Proc Natl Acad Sci U S A. 2005;102(43):15653–15658.
    1. Sequeira A, Mamdani F, Ernst C, et al. Global brain gene expression analysis links glutamatergic and GABAergic alterations to suicide and major depression. PloS One. 2009;4(8):e6585.
    1. Fatemi SH, Folsom TD, Rooney RJ, Thuras PD. Expression of GABAA α2-, β1- and ε-receptors are altered significantly in the lateral cerebellum of subjects with schizophrenia, major depression and bipolar disorder. Transl Psychiatry. 2013;3:e303.
    1. Klumpers UM, Veltman DJ, Drent ML, 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):565–574.
    1. Zhu H, Karolewicz B, Nail E, Stockmeier CA, Szebeni K, Ordway GA. Normal [3H]flunitrazepam binding to GABAA receptors in the locus coeruleus in major depression and suicide. Brain Res. 2006;1125(1):138–146.
    1. Stocks GM, Cheetham SC, Crompton MR, Katona CL, Horton RW. Benzodiazepine binding sites in amygdala and hippocampus of depressed suicide victims. J Affect Disord. 1990;18(1):11–15.
    1. Fatemi SH, Folsom TD, Thuras PD. Deficits in GABA(B) receptor system in schizophrenia and mood disorders: a postmortem study. Schizophr Res. 2011;128(1–3):37–43.
    1. Arranz B, Cowburn R, Eriksson A, Vestling M, Marcusson J. Gamma-aminobutyric acid-B (GABAB) binding sites in postmortem suicide brains. Neuropsychobiology. 1992;26(1–2):33–36.
    1. Cross JA, Cheetham SC, Crompton MR, Katona CL, Horton RW. Brain GABAB binding sites in depressed suicide victims. Psychiatry Res. 1988;26(2):119–129.
    1. Smith SS, Shen H, Gong QH, Zhou X. Neurosteroid regulation of GABA(A) receptors: focus on the α4 and δ subunits. Pharmacol Ther. 2007;116(1):58–76.
    1. Schmidt PJ, Nieman LK, Danaceau MA, Adams LF, Rubinow DR. Differential behavioral effects of gonadal steroids in women with and in those without premenstrual syndrome. N Engl J Med. 1998;338(4):209–216.
    1. Smith MJ, Adams LF, Schmidt PJ, Rubinow DR, Wassermann EM. Abnormal luteal phase excitability of the motor cortex in women with premenstrual syndrome. Biol Psychiatry. 2003;54(7):757–762.
    1. Majewska MD, Harrison NL, Schwartz RD, Barker JL, Paul SM. Steroid hormone metabolites are barbiturate-like modulators of the GABA receptor. Science. 1986;232(4753):1004–1007.
    1. Carboni E, Wieland S, Lan NC, Gee KW. Anxiolytic properties of endogenously occurring pregnanediols in two rodent models of anxiety. Psychopharmacology. 1996;126(2):173–178.
    1. Devaud LL, Purdy RH, Morrow AL. The neurosteroid, 3α-hydroxy-5α-pregnan-20-one, protects against bicuculline-induced seizures during ethanol withdrawal in rats. Alcohol Clin Exp Res. 1995;19(2):350–355.
    1. Shen H, Gong QH, Aoki C, et al. Reversal of neurosteroid effects at α4β2δ GABAA receptors triggers anxiety at puberty. Nat Neurosci. 2007;10(4):469–477.
    1. Smith SS, Ruderman Y, Frye C, Homanics G, Yuan M. Steroid withdrawal in the mouse results in anxiogenic effects of 3α,5β-THP: a possible model of premenstrual dysphoric disorder. Psychopharmacology. 2006;186(3):323–333.
    1. Wisden W, Laurie DJ, Monyer H, Seeburg PH. The distribution of 13 GABAA receptor subunit mRNAs in the rat brain. I. Telencephalon, diencephalon, mesencephalon. J Neurosci. 1992;12(3):1040–1062.
    1. Gulinello M, Gong QH, Li X, Smith SS. Short-term exposure to a neuroactive steroid increases α4 GABA(A) receptor subunit levels in association with increased anxiety in the female rat. Brain Res. 2001;910(1–2):55–66.
    1. Sundstrom-Poromaa I, Smith DH, Gong QH, et al. Hormonally regulated α(4)β(2)δ GABA(A) receptors are a target for alcohol. Nat Neurosci. 2002;5(8):721–722.
    1. Smith SS, Gong QH. Neurosteroid administration and withdrawal alter GABAA receptor kinetics in CA1 hippocampus of female rats. J Physiol. 2005;564(Pt 2):421–436.
    1. Smith SS, Gong QH, Li X, et al. Withdrawal from 3α-OH-5α-pregnan-20-one using a pseudopregnancy model alters the kinetics of hippocampal GABAA-gated current and increases the GABAA receptor α4 subunit in association with increased anxiety. J Neurosci. 1998;18(14):5275–5284.
    1. Maguire JL, Stell BM, Rafizadeh M, Mody I. Ovarian cycle-linked changes in GABA(A) receptors mediating tonic inhibition alter seizure susceptibility and anxiety. Nat Neurosci. 2005;8(6):797–804.
    1. Epperson CN, Haga K, Mason GF, et al. Cortical gamma-aminobutyric acid levels across the menstrual cycle in healthy women and those with premenstrual dysphoric disorder: a proton magnetic resonance spectroscopy study. Arch Gen Psychiatry. 2002;59(9):851–858.
    1. Uzunov DP, Cooper TB, Costa E, Guidotti A. Fluoxetine-elicited changes in brain neurosteroid content measured by negative ion mass fragmentography. Proc Natl Acad Sci U S A. 1996;93(22):12599–12604.
    1. Matsumoto K, Uzunova V, Pinna G, et al. Permissive role of brain allopregnanolone content in the regulation of pentobarbital-induced righting reflex loss. Neuropharmacology. 1999;38(7):955–963.
    1. Pehrson AL, Leiser SC, Gulinello M, et al. Treatment of cognitive dysfunction in major depressive disorder – a review of the preclinical evidence for efficacy of selective serotonin reuptake inhibitors, serotonin-norepinephrine reuptake inhibitors and the multimodal-acting antidepressant vortioxetine. Eur J Pharmacol. 2014 Aug 5; Epub.
    1. Meyer JH, Wilson AA, Sagrati S, et al. Serotonin transporter occupancy of five selective serotonin reuptake inhibitors at different doses: an [11C]DASB positron emission tomography study. Am J Psychiatry. 2004;161(5):826–835.
    1. Uzunova V, Sheline Y, Davis JM, et al. Increase in the cerebrospinal fluid content of neurosteroids in patients with unipolar major depression who are receiving fluoxetine or fluvoxamine. Proc Natl Acad Sci U S A. 1998;95(6):3239–3244.
    1. Romeo E, Ströhle A, Spalletta G, et al. Effects of antidepressant treatment on neuroactive steroids in major depression. Am J Psychiatry. 1998;155(7):910–913.
    1. Schule C, Baghai TC, Eser D, et al. Mirtazapine monotherapy versus combination therapy with mirtazapine and aripiprazole in depressed patients without psychotic features: a 4-week open-label parallel-group study. World J Biol Psychiatry. 2007;8(2):112–122.
    1. Birkenhäger TK, Moleman P, Nolen WA. Benzodiazepines for depression? A review of the literature. Int Clin Psychopharmacol. 1995;10(3):181–195.
    1. Petty F, Trivedi MH, Fulton M, Rush AJ. Benzodiazepines as antidepressants: does GABA play a role in depression? Biol Psychiatry. 1995;38(9):578–591.
    1. Fava M, McCall WV, Krystal A, et al. Eszopiclone co-administered with fluoxetine in patients with insomnia coexisting with major depressive disorder. Biol Psychiatry. 2006;59(11):1052–1060.
    1. Fava M, Schaefer K, Huang H, 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):473–479.
    1. El Zahaf NA, Salem Elhwuegi A. The effect of GABAmimetics on the duration of immobility in the forced swim test in albino mice. Libyan J Med. 2014;9:23480.
    1. Christensen T, Bétry C, Mnie-Filali O, et al. Synergistic antidepressant-like action of gaboxadol and escitalopram. Eur Neuropsychopharmacol. 2012;22(10):751–760.
    1. Kasper S, Ebert B, Larsen K, Tonnoir B. Combining escitalopram with gaboxadol provides no additional benefit in the treatment of patients with severe major depressive disorder. Int J Neuropsychopharmacol. 2012;15(6):715–725.
    1. Lambert MV, Robertson MM. Depression in epilepsy: etiology, phenomenology, and treatment. Epilepsia. 1999;40(Suppl 10):S21–S47.
    1. Brent DA, Crumrine PK, Varma RR, Allan M, Allman C. Phenobarbital treatment and major depressive disorder in children with epilepsy. Pediatrics. 1987;80(6):909–917.
    1. Mumford JP, Dam M. Meta-analysis of European placebo controlled studies of vigabatrin in drug resistant epilepsy. Br J Clin Pharmacol. 1989;27(Suppl 1):101S–107S.
    1. Levinson DF, Devinsky O. Psychiatric adverse events during vigabatrin therapy. Neurology. 1999;53(7):1503–1511.
    1. Ring HA, Crellin R, Kirker S, Reynolds EH. Vigabatrin and depression. J Neurol Neurosurg Psychiatry. 1993;56(8):925–928.
    1. Cryan JF, Slattery DA. GABAB receptors and depression. Current status. Adv Pharmacol. 2010;58:427–451.
    1. Post RM, Ketter TA, Joffe RT, Kramlinger KL. Lack of beneficial effects of l-baclofen in affective disorder. Int Clin Psychopharmacol. 1991;6(4):197–207.
    1. Mamelak M. Narcolepsy and depression and the neurobiology of gammahydroxybutyrate. Prog Neurobiol. 2009;89(2):193–219.
    1. Chouinard G. Issues in the clinical use of benzodiazepines: potency, withdrawal, and rebound. J Clin Psychiatry. 2004;65(Suppl 5):7–12.
    1. Pietrzak RH, Scott JC, Harel BT, Lim YY, Snyder PJ, Maruff P. A process-based approach to characterizing the effect of acute alprazolam challenge on visual paired associate learning and memory in healthy older adults. Hum Psychopharmacol. 2012;27(6):549–558.
    1. Lüscher C, Jan LY, Stoffel M, Malenka RC, Nicoll RA. G protein-coupled inwardly rectifying K+ channels (GIRKs) mediate postsynaptic but not presynaptic transmitter actions in hippocampal neurons. Neuron. 1997;19(3):687–695.
    1. Aznar S, Qian Z, Shah R, Rahbek B, Knudsen GM. The 5-HT1A serotonin receptor is located on calbindin- and parvalbumin-containing neurons in the rat brain. Brain Res. 2003;959(1):58–67.
    1. Levkovitz Y, Segal M. Serotonin 5-HT1A receptors modulate hippocampal reactivity to afferent stimulation. J Neurosci. 1997;17(14):5591–5598.
    1. Lladó-Pelfort L, Assié MB, Newman-Tancredi A, Artigas F, Celada P. In vivo electrophysiological and neurochemical effects of the selective 5-HT1A receptor agonist, F13640, at pre- and postsynaptic 5-HT1A receptors in the rat. Psychopharmacology. 2012;221(2):261–272.
    1. Lladó-Pelfort L, Santana N, Ghisi V, Artigas F, Celada P. 5-HT1A receptor agonists enhance pyramidal cell firing in prefrontal cortex through a preferential action on GABA interneurons. Cereb Cortex. 2012;22(7):1487–1497.
    1. Bombardi C, Di Giovanni G. Functional anatomy of 5-HT2A receptors in the amygdala and hippocampal complex: relevance to memory functions. Exp Brain Res. 2013;230(4):427–439.
    1. López-Giménez JF, Vilaró MT, Palacios JM, Mengod G. Mapping of 5-HT2A receptors and their mRNA in monkey brain: [3H] MDL100,907 autoradiography and in situ hybridization studies. J Comp Neurol. 2001;429(4):571–589.
    1. Celada P, Puig MV, Artigas F. Serotonin modulation of cortical neurons and networks. Front Integr Neurosci. 2013;7:25.
    1. Lüttgen M, Ove Ogren S, Meister B. Chemical identity of 5-HT2A receptor immunoreactive neurons of the rat septal complex and dorsal hippocampus. Brain Res. 2004;1010(1–2):156–165.
    1. Puig MV, Celada P, Diaz-Mataix L, Artigas F. In vivo modulation of the activity of pyramidal neurons in the rat medial prefrontal cortex by 5-HT2A receptors: relationship to thalamocortical afferents. Cereb Cortex. 2003;13(8):870–882.
    1. Wood J, Kim Y, Moghaddam B. Disruption of prefrontal cortex large scale neuronal activity by different classes of psychotomimetic drugs. J Neurosci. 2012;32(9):3022–3031.
    1. Ashby CR, Jr, Jiang LH, Kasser RJ, Wang RY. Electrophysiological characterization of 5-hydroxytryptamine2 receptors in the rat medial prefrontal cortex. J Pharmacol Exp Ther. 1990;252(1):171–178.
    1. Wang RY, Arvanov VL. M100907, a highly selective 5-HT2A receptor antagonist and a potential atypical antipsychotic drug, facilitates induction of long-term potentiation in area CA1 of the rat hippocampal slice. Brain Res. 1998;779(1–2):309–313.
    1. Bloom FE, Morales M. The central 5-HT3 receptor in CNS disorders. Neurochem Res. 1998;23(5):653–659.
    1. Morales M, Bloom FE. The 5-HT3 receptor is present in different subpopulations of GABAergic neurons in the rat telencephalon. J Neurosci. 1997;17(9):3157–3167.
    1. Puig MV, Santana N, Celada P, Mengod G, Artigas F. In vivo excitation of GABA interneurons in the medial prefrontal cortex through 5-HT3 receptors. Cereb Cortex. 2004;14(12):1365–1375.
    1. Ashby CR, Jr, Minabe Y, Edwards E, Wang RY. 5-HT3-like receptors in the rat medial prefrontal cortex: an electrophysiological study. Brain Res. 1991;550(2):181–191.
    1. Reznic J, Staubli U. Effects of 5-HT3 receptor antagonism on hippocampal cellular activity in the freely moving rat. J Neurophysiol. 1997;77(1):517–521.
    1. Stäubli U, Xu FB. Effects of 5-HT3 receptor antagonism on hippocampal theta rhythm, memory, and LTP induction in the freely moving rat. J Neurosci. 1995;15(3 Pt 2):2445–2452.
    1. Chapin EM, Haj-Dahmane S, Torres G, Andrade R. The 5-HT(4) receptor-induced depolarization in rat hippocampal neurons is mediated by cAMP but is independent of I(h) Neurosci Lett. 2002;324(1):1–4.
    1. Waeber C, Sebben M, Nieoullon A, Bockaert J, Dumuis A. Regional distribution and ontogeny of 5-HT4 binding sites in rodent brain. Neuropharmacology. 1994;33(3–4):527–541.
    1. Bianchi C, Rodi D, Marino S, Beani L, Siniscalchi A. Dual effects of 5-HT4 receptor activation on GABA release from guinea pig hippocampal slices. Neuroreport. 2002;13(17):2177–2180.
    1. Cai X, Flores-Hernandez J, Feng J, Yan Z. Activity-dependent bidirectional regulation of GABA(A) receptor channels by the 5-HT(4) receptor-mediated signalling in rat prefrontal cortical pyramidal neurons. J Physiol. 2002;540(Pt 3):743–759.
    1. Francken BJ, Jurzak M, Vanhauwe JF, Luyten WH, Leysen JE. The human 5-HT5A receptor couples to Gi/Go proteins and inhibits adenylate cyclase in HEK 293 cells. Eur J Pharmacol. 1998;361(2–3):299–309.
    1. Goodfellow NM, Bailey CD, Lambe EK. The native serotonin 5-HT(5A) receptor: electrophysiological characterization in rodent cortex and 5-HT(1A)-mediated compensatory plasticity in the knockout mouse. J Neurosci. 2012;32(17):5804–5809.
    1. Oliver KR, Kinsey AM, Wainwright A, Sirinathsinghji DJ. Localization of 5-HT(5A) receptor-like immunoreactivity in the rat brain. Brain Res. 2000;867(1–2):131–142.
    1. Ruat M, Traiffort E, Arrang JM, et al. A novel rat serotonin (5-HT6) receptor: molecular cloning, localization and stimulation of cAMP accumulation. Biochem Biophys Res Commun. 1993;193(1):268–276.
    1. Gerard C, Martres MP, Lefevre K, et al. Immuno-localization of serotonin 5-HT6 receptor-like material in the rat central nervous system. Brain Res. 1997;746(1–2):207–219.
    1. West PJ, Marcy VR, Marino MJ, Schaffhauser H. Activation of the 5-HT(6) receptor attenuates long-term potentiation and facilitates GABAergic neurotransmission in rat hippocampus. Neuroscience. 2009;164(2):692–701.
    1. Schechter LE, Lin Q, Smith DL, et al. Neuropharmacological profile of novel and selective 5-HT6 receptor agonists: WAY-181187 and WAY-208466. Neuropsychopharmacology. 2008;33(6):1323–1335.
    1. Woolley ML, Marsden CA, Fone KC. 5-HT6 receptors. Curr Drug Targets CNS Neurol Disord. 2004;3(1):59–79.
    1. Marazziti D, Baroni S, Pirone A, et al. Distribution of serotonin receptor of type 6 (5-HT(6)) in human brain post-mortem. A pharmacology, autoradiography and immunohistochemistry study. Neurochem Res. 2012;37(5):920–927.
    1. Marazziti D, Baroni S, Pirone A, et al. Serotonin receptor of type 6 (5-HT6) in human prefrontal cortex and hippocampus post-mortem: an immunohistochemical and immunofluorescence study. Neurochem Int. 2013;62(2):182–188.
    1. Hedlund PB, Sutcliffe JG. Functional, molecular and pharmacological advances in 5-HT7 receptor research. Trends Pharmacol Sci. 2004;25(9):481–486.
    1. Neumaier JF, Sexton TJ, Yracheta J, Diaz AM, Brownfield M. Localization of 5-HT(7) receptors in rat brain by immunocytochemistry, in situ hybridization, and agonist stimulated cFos expression. J Chem Neuroanat. 2001;21(1):63–73.
    1. Geurts FJ, De Schutter E, Timmermans JP. Localization of 5-HT2A, 5-HT3, 5-HT5A and 5-HT7 receptor-like immunoreactivity in the rat cerebellum. J Chem Neuroanat. 2002;24(1):65–74.
    1. Tokarski K, Kusek M, Hess G. 5-HT7 receptors modulate GABAergic transmission in rat hippocampal CA1 area. J Physiol Pharmacol. 2011;62(5):535–540.
    1. Bacon WL, Beck SG. 5-Hydroxytryptamine(7) receptor activation decreases slow afterhyperpolarization amplitude in CA3 hippocampal pyramidal cells. J Pharmacol Exp Ther. 2000;294(2):672–679.
    1. Fan LL, Zhang QJ, Liu J, et al. In vivo effect of 5-HT(7) receptor agonist on pyramidal neurons in medial frontal cortex of normal and 6-hydroxydopamine-lesioned rats: an electrophysiological study. Neuroscience. 2011;190:328–338.
    1. Owens MJ, Morgan WN, Plott SJ, Nemeroff CB. Neurotransmitter receptor and transporter binding profile of antidepressants and their metabolites. J Pharmacol Exp Ther. 1997;283(3):1305–1322.
    1. Zhong P, Yan Z. Differential regulation of the excitability of prefrontal cortical fast-spiking interneurons and pyramidal neurons by serotonin and fluoxetine. PloS One. 2011;6(2):e16970.
    1. Méndez P, Pazienti A, Szabó G, Bacci A. Direct alteration of a specific inhibitory circuit of the hippocampus by antidepressants. J Neurosci. 2012;32(47):16616–16628.
    1. Gören MZ, Küçükibrahimoglu E, Berkman K, Terzioglu B. Fluoxetine partly exerts its actions through GABA: a neurochemical evidence. Neurochem Res. 2007;32(9):1559–1565.
    1. Robinson RT, Drafts BC, Fisher JL. Fluoxetine increases GABA(A) receptor activity through a novel modulatory site. J Pharmacol Exp Ther. 2003;304(3):978–984.
    1. Derry JM, Paulsen IM, Davies M, Dunn SM. A single point mutation of the GABA(A) receptor α5-subunit confers fluoxetine sensitivity. Neuropharmacology. 2007;52(2):497–505.
    1. Ye ZY, Zhou KQ, Xu TL, Zhou JN. Fluoxetine potentiates GABAergic IPSCs in rat hippocampal neurons. Neurosci Lett. 2008;442(1):24–29.
    1. Henry ME, Schmidt ME, Hennen J, et al. A comparison of brain and serum pharmacokinetics of R-fluoxetine and racemic fluoxetine: a 19-F MRS study. Neuropsychopharmacology. 2005;30(8):1576–1583.
    1. Liu X, Smith BJ, Chen C, et al. Evaluation of cerebrospinal fluid concentration and plasma free concentration as a surrogate measurement for brain free concentration. Drug Metab Dispos. 2006;34(9):1443–1447.
    1. Mårtensson B, Nyberg S, Toresson G, Brodin E, Bertilsson L. Fluoxetine treatment of depression. Clinical effects, drug concentrations and monoamine metabolites and N-terminally extended substance P in cerebrospinal fluid. Acta Psychiatr Scand. 1989;79(6):586–596.
    1. Zhong P, Yan Z. Chronic antidepressant treatment alters serotonergic regulation of GABA transmission in prefrontal cortical pyramidal neurons. Neuroscience. 2004;129(1):65–73.
    1. Khisti RT, Chopde CT, Jain SP. Antidepressant-like effect of the neurosteroid 3α-hydroxy-5α-pregnan-20-one in mice forced swim test. Pharmacol Biochem Behav. 2000;67(1):137–143.
    1. Pehrson AL, Sanchez C. Serotonergic modulation of glutamate neurotransmission as a strategy for treating depression and cognitive dysfunction. CNS Spectr. 2014;19(2):121–133.
    1. Westrich L, Pehrson A, Zhong H, et al. In vitro and in vivo effects of the multimodal antidepressant vortioxetine (Lu AA21004) at human and rat targets. Int J Psychiatry Clin Pract. 2012;16(Suppl 1):47.
    1. Dale E, Zhang H, Leiser SC, et al. Vortioxetine disinhibits pyramidal cell function and enhances synaptic plasticity in the rat hippocampus. J Psychopharmacol. 2014;28(10):891–902.
    1. Riga MS, Celada P, Sanchez C, Artigas F. Role of 5-HT3 receptors in the mechanism of action of the investigational antidepressant vortioxetine. Eur Neuropsychopharmacol. 2013;23(Suppl 2):S393–S394.
    1. Leiser SC, Pehrson AL, Robichaud PJ, Sanchez C. The multimodal antidepressant vortioxetine increases frontal cortical oscillations unlike escitalopram and duloxetine – a quantitative electroencephalographic study in the rat. Br J Pharmacol. 2014 May 21; Epub.
    1. Alvarez E, Perez V, Artigas F. Pharmacology and clinical potential of vortioxetine in the treatment of major depressive disorder. Neuropsychiatric Dis Treat. 2014;10:1297–1307.

Source: PubMed

3
Tilaa