New perspectives on the involvement of mTOR in depression as well as in the action of antidepressant drugs.

Despite the revolution in recent decades regarding monoamine involvement in the management of major depressive disorder (MDD), the biological mechanisms underlying this psychiatric disorder are still poorly understood. Currently available treatments require long time courses to establish antidepressant response and a significant percentage of people are refractory to single drug or combination drug treatment. These issues, and recent findings demonstrating the involvement of synaptic plasticity in the pathophysiological mechanisms of MDD, are encouraging researchers to explore the molecular mechanisms underlying psychiatric disease in more depth. The discovery of the rapid antidepressant effect exerted by glutamatergic and cholinergic agents highlights the mammalian target of rapamycin (mTOR) pathway as a critical pathway that contributes to the efficacy of these pharmacological agents in clinical and pre-clinical research. The mTOR pathway is a downstream intracellular signal that transmits information after the direct activation of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) and neurotrophic factor receptors. Activation of these receptors is hypothesized to be one of the major axes involved in the synthesis of synaptogenic proteins underlying synaptic plasticity and critical to both the rapid and delayed effects exerted by classic antidepressants. This review focuses on the involvement of mTOR in the pathophysiology of depression and on molecular mechanisms involved in the activity of emerging and classic antidepressant agents.

[1]  A. Yusuf mTOR signaling in growth and metabolism , 2017 .

[2]  Oliver H. Miller,et al.  Two cellular hypotheses explaining the initiation of ketamine's antidepressant actions: Direct inhibition and disinhibition , 2016, Neuropharmacology.

[3]  J. Duan,et al.  Involvement of normalized NMDA receptor and mTOR-related signaling in rapid antidepressant effects of Yueju and ketamine on chronically stressed mice , 2015, Scientific Reports.

[4]  S. Deutsch,et al.  NMDA receptor activation regulates sociability by its effect on mTOR signaling activity , 2015, Progress in Neuro-Psychopharmacology and Biological Psychiatry.

[5]  M. Jou,et al.  AMPA Receptor–mTOR Activation is Required for the Antidepressant-Like Effects of Sarcosine during the Forced Swim Test in Rats: Insertion of AMPA Receptor may Play a Role , 2015, Front. Behav. Neurosci..

[6]  Young Hoon Kim,et al.  Differential effects of antidepressant drugs on mTOR signalling in rat hippocampal neurons. , 2014, The international journal of neuropsychopharmacology.

[7]  E. Delpire,et al.  GluN2B-containing NMDA receptors regulate depression-like behavior and are critical for the rapid antidepressant actions of ketamine , 2014, eLife.

[8]  H. Abelaira,et al.  Epigenetic and epistatic interactions between serotonin transporter and brain-derived neurotrophic factor genetic polymorphism: Insights in depression , 2014, Neuroscience.

[9]  Zhi-qiang Zhou,et al.  Ketamine-induced antidepressant effects are associated with AMPA receptors-mediated upregulation of mTOR and BDNF in rat hippocampus and prefrontal cortex , 2014, European Psychiatry.

[10]  K. Maiese Driving neural regeneration through the mammalian target of rapamycin , 2014, Neural regeneration research.

[11]  H. Nawa,et al.  mTOR signaling and its roles in normal and abnormal brain development , 2014, Front. Mol. Neurosci..

[12]  H. Abelaira,et al.  The role of mTOR in depression and antidepressant responses. , 2014, Life sciences.

[13]  G. Aghajanian,et al.  REDD1 is essential for stress-induced synaptic loss and depressive behavior , 2014, Nature Medicine.

[14]  J. Long,et al.  Monoacylglycerol Lipase Inhibition Blocks Chronic Stress-Induced Depressive-Like Behaviors via Activation of mTOR Signaling , 2014, Neuropsychopharmacology.

[15]  Nanxin Li,et al.  Scopolamine Rapidly Increases Mammalian Target of Rapamycin Complex 1 Signaling, Synaptogenesis, and Antidepressant Behavioral Responses , 2013, Biological Psychiatry.

[16]  Hai-shui Shi,et al.  Glycine site N-methyl-D-aspartate receptor antagonist 7-CTKA produces rapid antidepressant-like effects in male rats. , 2013, Journal of psychiatry & neuroscience : JPN.

[17]  Dennis S. Charney,et al.  Rapid and Longer-Term Antidepressant Effects of Repeated Ketamine Infusions in Treatment-Resistant Major Depression , 2013, Biological Psychiatry.

[18]  D. Inta,et al.  Pharmacological blockade of GluN2B-containing NMDA receptors induces antidepressant-like effects lacking psychotomimetic action and neurotoxicity in the perinatal and adult rodent brain , 2013, Progress in Neuro-Psychopharmacology and Biological Psychiatry.

[19]  Young Hoon Kim,et al.  Effect of treadmill exercise on the BDNF-mediated pathway in the hippocampus of stressed rats , 2013, Neuroscience Research.

[20]  Jin-Hui Wang,et al.  The Stability of NR2B in the Nucleus Accumbens Controls Behavioral and Synaptic Adaptations to Chronic Stress , 2013, Biological Psychiatry.

[21]  M. Furey,et al.  Antidepressant Effects of the Muscarinic Cholinergic Receptor Antagonist Scopolamine: A Review , 2013, Biological Psychiatry.

[22]  R. Duman,et al.  Activation of Mammalian Target of Rapamycin and Synaptogenesis: Role in the Actions of Rapid-Acting Antidepressants , 2013, Biological Psychiatry.

[23]  Y. Tizabi,et al.  Antidepressant effects of AMPA and ketamine combination: role of hippocampal BDNF, synapsin, and mTOR , 2013, Psychopharmacology.

[24]  G. Aghajanian,et al.  GSK-3 Inhibition Potentiates the Synaptogenic and Antidepressant-Like Effects of Subthreshold Doses of Ketamine , 2013, Neuropsychopharmacology.

[25]  H. Abelaira,et al.  Imipramine reverses alterations in cytokines and BDNF levels induced by maternal deprivation in adult rats , 2013, Behavioural Brain Research.

[26]  H. Abelaira,et al.  Effects of lamotrigine on behavior, oxidative parameters and signaling cascades in rats exposed to the chronic mild stress model , 2013, Neuroscience Research.

[27]  Zhi-qiang Zhou,et al.  Acute administration of ketamine in rats increases hippocampal BDNF and mTOR levels during forced swimming test , 2013, Upsala journal of medical sciences.

[28]  M. Austin,et al.  Reduced phosphorylation of the mTOR signaling pathway components in the amygdala of rats exposed to chronic stress , 2013, Progress in Neuro-Psychopharmacology and Biological Psychiatry.

[29]  Jean-Marie C. Bouteiller,et al.  Roles of group I metabotropic glutamate receptors under physiological conditions and in neurodegeneration , 2012 .

[30]  Zhi-qiang Zhou,et al.  Tramadol Pretreatment Enhances Ketamine-Induced Antidepressant Effects and Increases Mammalian Target of Rapamycin in Rat Hippocampus and Prefrontal Cortex , 2012, Journal of biomedicine & biotechnology.

[31]  D. Sabatini,et al.  mTOR Signaling in Growth Control and Disease , 2012, Cell.

[32]  Nanxin Li,et al.  Signaling pathways underlying the rapid antidepressant actions of ketamine , 2012, Neuropharmacology.

[33]  S. Chaki,et al.  Involvement of AMPA receptor in both the rapid and sustained antidepressant-like effects of ketamine in animal models of depression , 2011, Behavioural Brain Research.

[34]  M. Austin,et al.  The mTOR signaling pathway in the prefrontal cortex is compromised in major depressive disorder , 2011, Progress in Neuro-Psychopharmacology and Biological Psychiatry.

[35]  E. Kavalali,et al.  NMDA Receptor Blockade at Rest Triggers Rapid Behavioural Antidepressant Responses , 2011, Nature.

[36]  A. Mallinger,et al.  Rapid decrease in depressive symptoms with an N-methyl-d-aspartate antagonist in ECT-resistant major depression , 2011, Progress in Neuro-Psychopharmacology and Biological Psychiatry.

[37]  A. Pillai,et al.  Long-Term Continuous Corticosterone Treatment Decreases VEGF Receptor-2 Expression in Frontal Cortex , 2011, PloS one.

[38]  R. Jope,et al.  Inhibition of glycogen synthase kinase-3 is necessary for the rapid antidepressant effect of ketamine in mice , 2011, Molecular Psychiatry.

[39]  Nanxin Li,et al.  Glutamate N-methyl-D-aspartate Receptor Antagonists Rapidly Reverse Behavioral and Synaptic Deficits Caused by Chronic Stress Exposure , 2011, Biological Psychiatry.

[40]  Mesut Çetin,et al.  Beyond the glutamate N-methyl D-aspartate receptor in major depressive disorder: the mTOR signaling pathway / Majör depresif bozuklukta glutamat N-metil-D-aspartat reseptörlerinin ötesi: mTOR sinyal yolağı , 2011 .

[41]  Brian K. Kennedy,et al.  TOR on the brain , 2011, Experimental Gerontology.

[42]  D. Sabatini,et al.  mTOR: from growth signal integration to cancer, diabetes and ageing , 2010, Nature Reviews Molecular Cell Biology.

[43]  Ronald S Duman,et al.  Peripheral BDNF Produces Antidepressant-Like Effects in Cellular and Behavioral Models , 2011, Neuropsychopharmacology.

[44]  I. Lucki,et al.  Fluoxetine treatment induces dose dependent alterations in depression associated behavior and neural plasticity in female mice , 2010, Neuroscience Letters.

[45]  S. Horvath,et al.  Biomarkers to Predict Antidepressant Response , 2010, Current psychiatry reports.

[46]  Nanxin Li,et al.  mTOR-Dependent Synapse Formation Underlies the Rapid Antidepressant Effects of NMDA Antagonists , 2010, Science.

[47]  D. Luckenbaugh,et al.  A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. , 2010, Archives of general psychiatry.

[48]  M. Furey,et al.  Replication of Scopolamine's Antidepressant Efficacy in Major Depressive Disorder: A Randomized, Placebo-Controlled Clinical Trial , 2010, Biological Psychiatry.

[49]  E. Klann,et al.  mTOR signaling: At the crossroads of plasticity, memory and disease , 2010, Trends in Neurosciences.

[50]  J. R. Gomes,et al.  BDNF-induced changes in the expression of the translation machinery in hippocampal neurons: protein levels and dendritic mRNA. , 2009, Journal of proteome research.

[51]  M. Kawamura,et al.  Brain-derived Neurotrophic Factor Enhances the Basal Rate of Protein Synthesis by Increasing Active Eukaryotic Elongation Factor 2 Levels and Promoting Translation Elongation in Cortical Neurons* , 2009, The Journal of Biological Chemistry.

[52]  M. Krams,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 , 2008, Journal of clinical psychopharmacology.

[53]  A. Zangen,et al.  Age‐dependent effects of chronic stress on brain plasticity and depressive behavior , 2008, Journal of neurochemistry.

[54]  H. Schmidt,et al.  Future Antidepressant Targets: Neurotrophic Factors and Related Signaling Cascades. , 2008, Drug discovery today. Therapeutic strategies.

[55]  H. Manji,et al.  The Role of AMPA receptor modulation in the treatment of neuropsychiatric diseases , 2008, Experimental Neurology.

[56]  Carlos A. Zarate,et al.  Targeting the glutamatergic system to develop novel, improved therapeutics for mood disorders , 2008, Nature Reviews Drug Discovery.

[57]  June-Seek Choi,et al.  Vascular endothelial growth factor (VEGF) signaling regulates hippocampal neurons by elevation of intracellular calcium and activation of calcium/calmodulin protein kinase II and mammalian target of rapamycin. , 2008, Cellular signalling.

[58]  Guang Chen,et al.  Cellular Mechanisms Underlying the Antidepressant Effects of Ketamine: Role of α-Amino-3-Hydroxy-5-Methylisoxazole-4-Propionic Acid Receptors , 2008, Biological Psychiatry.

[59]  B. Moghaddam,et al.  NMDA Receptor Hypofunction Produces Opposite Effects on Prefrontal Cortex Interneurons and Pyramidal Neurons , 2007, The Journal of Neuroscience.

[60]  H. Manji,et al.  The Anticonvulsants Lamotrigine, Riluzole, and Valproate Differentially Regulate AMPA Receptor Membrane Localization: Relationship to Clinical Effects in Mood Disorders , 2007, Neuropsychopharmacology.

[61]  R. Duman,et al.  VEGF is an essential mediator of the neurogenic and behavioral actions of antidepressants , 2007, Proceedings of the National Academy of Sciences.

[62]  A. Serretti,et al.  Meta-analysis of serotonin transporter gene promoter polymorphism (5-HTTLPR) association with selective serotonin reuptake inhibitor efficacy in depressed patients , 2007, Molecular Psychiatry.

[63]  G. Collingridge,et al.  LTP Inhibits LTD in the Hippocampus via Regulation of GSK3β , 2007, Neuron.

[64]  S. Snyder,et al.  The Cationic Amino Acid Transporters CAT1 and CAT3 Mediate NMDA Receptor Activation-Dependent Changes in Elaboration of Neuronal Processes via the Mammalian Target of Rapamycin mTOR Pathway , 2007, The Journal of Neuroscience.

[65]  A. Malafosse,et al.  Alteration in Kinase Activity But Not in Protein Levels of Protein Kinase B and Glycogen Synthase Kinase-3β in Ventral Prefrontal Cortex of Depressed Suicide Victims , 2007, Biological Psychiatry.

[66]  Debabrata Panja,et al.  Dual regulation of translation initiation and peptide chain elongation during BDNF‐induced LTP in vivo: evidence for compartment‐specific translation control , 2006, Journal of neurochemistry.

[67]  C. Siao,et al.  Genetic Variant BDNF (Val66Met) Polymorphism Alters Anxiety-Related Behavior , 2006, Science.

[68]  W. Sossin,et al.  Serotonin Increases Phosphorylation of Synaptic 4EBP through TOR, but Eukaryotic Initiation Factor 4E Levels Do Not Limit Somatic Cap-Dependent Translation in Aplysia Neurons , 2006, Molecular and Cellular Biology.

[69]  Ming You,et al.  TSC2 Integrates Wnt and Energy Signals via a Coordinated Phosphorylation by AMPK and GSK3 to Regulate Cell Growth , 2006, Cell.

[70]  L. Barrier,et al.  Group I metabotropic glutamate receptors activate the p70S6 kinase via both mammalian target of rapamycin (mTOR) and extracellular signal-regulated kinase (ERK 1/2) signaling pathways in rat striatal and hippocampal synaptoneurosomes , 2006, Neurochemistry International.

[71]  Paul J Carlson,et al.  A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. , 2006, Archives of general psychiatry.

[72]  R. Duman,et al.  A Neurotrophic Model for Stress-Related Mood Disorders , 2006, Biological Psychiatry.

[73]  C. Bramham,et al.  Chronic fluoxetine induces region‐specific changes in translation factor eIF4E and eEF2 activity in the rat brain , 2006, The European journal of neuroscience.

[74]  B. Lebowitz,et al.  Medication augmentation after the failure of SSRIs for depression. , 2006, The New England journal of medicine.

[75]  M. Hall,et al.  TOR Signaling in Growth and Metabolism , 2006, Cell.

[76]  M. Furey,et al.  Antidepressant Efficacy of the Antimuscarinic Drug Scopolamine , 2006 .

[77]  C. Hoogenraad,et al.  Control of Dendritic Arborization by the Phosphoinositide-3′-Kinase–Akt–Mammalian Target of Rapamycin Pathway , 2005, The Journal of Neuroscience.

[78]  Gang-yi Wu,et al.  Regulation of Dendritic Morphogenesis by Ras–PI3K–Akt–mTOR and Ras–MAPK Signaling Pathways , 2005, The Journal of Neuroscience.

[79]  N. Sonenberg,et al.  The Translation Repressor 4E-BP2 Is Critical for eIF4F Complex Formation, Synaptic Plasticity, and Memory in the Hippocampus , 2005, The Journal of Neuroscience.

[80]  Kenta Hara,et al.  Brain-Derived Neurotrophic Factor Induces Mammalian Target of Rapamycin-Dependent Local Activation of Translation Machinery and Protein Synthesis in Neuronal Dendrites , 2004, The Journal of Neuroscience.

[81]  A. Schatzberg,et al.  Effects of the serotonin transporter gene promoter polymorphism on mirtazapine and paroxetine efficacy and adverse events in geriatric major depression. , 2004, Archives of general psychiatry.

[82]  W. Sossin,et al.  5‐HT stimulates eEF2 dephosphorylation in a rapamycin‐sensitive manner in Aplysia neurites , 2004, Journal of neurochemistry.

[83]  J Ormel,et al.  Functional disability and depression in the general population. Results from the Netherlands Mental Health Survey and Incidence Study (NEMESIS) , 2004, Acta psychiatrica Scandinavica.

[84]  M. Barrot,et al.  Essential role of brain-derived neurotrophic factor in adult hippocampal function. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[85]  J. Blenis,et al.  Target of rapamycin (TOR): an integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression , 2004, Oncogene.

[86]  Christopher G. Proud,et al.  A Novel mTOR-Regulated Phosphorylation Site in Elongation Factor 2 Kinase Modulates the Activity of the Kinase and Its Binding to Calmodulin , 2004, Molecular and Cellular Biology.

[87]  J. Medina,et al.  ERK1/2 activation is necessary for BDNF to increase dendritic spine density in hippocampal CA1 pyramidal neurons. , 2004, Learning & memory.

[88]  Olga V. Demler,et al.  The epidemiology of major depressive disorder: results from the National Comorbidity Survey Replication (NCS-R). , 2003, JAMA.

[89]  A. Nairn,et al.  Adenylyl cyclase-dependent form of chemical long-term potentiation triggers translational regulation at the elongation step , 2003, Neuroscience.

[90]  E. Castrén,et al.  Activation of the TrkB Neurotrophin Receptor Is Induced by Antidepressant Drugs and Is Required for Antidepressant-Induced Behavioral Effects , 2003, The Journal of Neuroscience.

[91]  M. Cuesta,et al.  [Neurobiology of depression]. , 2002, Anales del sistema sanitario de Navarra.

[92]  R. Duman,et al.  Brain-Derived Neurotrophic Factor Produces Antidepressant Effects in Behavioral Models of Depression , 2002, The Journal of Neuroscience.

[93]  Mark Farrant,et al.  NMDA receptor subunits: diversity, development and disease , 2001, Current Opinion in Neurobiology.

[94]  Jeffrey A Lieberman,et al.  Effects of Ketamine, MK-801, and Amphetamine on Regional Brain 2-Deoxyglucose Uptake in Freely Moving Mice , 2000, Neuropsychopharmacology.

[95]  John H Krystal,et al.  Antidepressant effects of ketamine in depressed patients , 2000, Biological Psychiatry.

[96]  A. Gingras,et al.  μ-Opioid Receptor Activates Signaling Pathways Implicated in Cell Survival and Translational Control* , 1998, The Journal of Biological Chemistry.

[97]  J. Heitman,et al.  Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast , 1991, Science.