Rapid-acting antidepressants.
暂无分享,去创建一个
[1] R. McIntyre,et al. Pharmacological interventions targeting anhedonia in patients with major depressive disorder: A systematic review , 2019, Progress in Neuro-Psychopharmacology and Biological Psychiatry.
[2] J. Callanan,et al. Comparison of electroencephalogram (EEG) response to MDPV versus the hallucinogenic drugs MK-801 and ketamine in rats , 2019, Experimental Neurology.
[3] K. Hashimoto,et al. Increased expression of inwardly rectifying Kir4.1 channel in the parietal cortex from patients with major depressive disorder. , 2019, Journal of affective disorders.
[4] S. Wilkinson,et al. A new generation of antidepressants: an update on the pharmaceutical pipeline for novel and rapid-acting therapeutics in mood disorders based on glutamate/GABA neurotransmitter systems. , 2019, Drug discovery today.
[5] S. Dursun,et al. Ayahuasca: Psychological and Physiologic Effects, Pharmacology and Potential Uses in Addiction and Mental Illness , 2019, Current neuropharmacology.
[6] Sara Mohamed,et al. Sex Differences in the Temporal Neuromolecular and Synaptogenic Effects of the Rapid-acting Antidepressant Drug Ketamine in the Mouse Brain , 2019, Neuroscience.
[7] K. Hashimoto,et al. Lack of rapid antidepressant effects of Kir4.1 channel inhibitors in a chronic social defeat stress model: Comparison with (R)-ketamine , 2019, Pharmacology Biochemistry and Behavior.
[8] Zhongming Chen,et al. BDNF mediates the protective effects of scopolamine in reserpine-induced depression-like behaviors via up-regulation of 5-HTT and TPH1 , 2019, Psychiatry Research.
[9] A. Nugent,et al. Synaptic potentiation and rapid antidepressant response to ketamine in treatment-resistant major depression: A replication study , 2019, Psychiatry Research: Neuroimaging.
[10] M. Banks,et al. Effects of acute and repeated treatment with serotonin 5-HT2A receptor agonist hallucinogens on intracranial self-stimulation in rats. , 2019, Experimental and clinical psychopharmacology.
[11] S. Thompson,et al. Molecular Pharmacology and Neurobiology of Rapid-Acting Antidepressants. , 2019, Annual review of pharmacology and toxicology.
[12] K. Hashimoto,et al. An update on ketamine and its two enantiomers as rapid-acting antidepressants , 2018, Expert review of neurotherapeutics.
[13] S. Chaki,et al. Is the History Repeated? Can (2R,6R)-Hydroxynorketamine be Another Antidepressant? , 2018, Journal of experimental neuroscience.
[14] F. McMahon,et al. Exploratory genome-wide association analysis of response to ketamine and a polygenic analysis of response to scopolamine in depression , 2018, Translational Psychiatry.
[15] T. Gould,et al. Mouse, rat, and dog bioavailability and mouse oral antidepressant efficacy of (2R,6R)-hydroxynorketamine , 2018, Journal of psychopharmacology.
[16] C. Zarate,et al. Glutamatergic Neurotransmission: Pathway to Developing Novel Rapid-Acting Antidepressant Treatments , 2018, The international journal of neuropsychopharmacology.
[17] Dan T. Rosenbaum,et al. Microdosing psychedelics: personality, mental health, and creativity differences in microdosers , 2018, Psychopharmacology.
[18] Matthew W. Johnson,et al. The abuse potential of medical psilocybin according to the 8 factors of the Controlled Substances Act , 2018, Neuropharmacology.
[19] K. Hashimoto,et al. Comparison of rapid and long-lasting antidepressant effects of negative modulators of α5-containing GABAA receptors and (R)‑ketamine in a chronic social defeat stress model , 2018, Pharmacology Biochemistry and Behavior.
[20] A. Yli-Hankala,et al. Cortical Excitability and Activation of TrkB Signaling During Rebound Slow Oscillations Are Critical for Rapid Antidepressant Responses , 2018, Molecular Neurobiology.
[21] J. Krystal,et al. The neurobiology of depression, ketamine and rapid-acting antidepressants: Is it glutamate inhibition or activation? , 2018, Pharmacology & therapeutics.
[22] K. Nakazawa,et al. AMPA Receptor Activation–Independent Antidepressant Actions of Ketamine Metabolite (S)-Norketamine , 2018, Biological Psychiatry.
[23] Andrzej Pilc,et al. The potential antidepressant action and adverse effects profile of scopolamine co-administered with the mGlu7 receptor allosteric agonist AMN082 in mice , 2018, Neuropharmacology.
[24] P. Witt-Enderby,et al. DARK Classics in Chemical Neuroscience: Ibogaine. , 2018, ACS chemical neuroscience.
[25] M. Rasenick,et al. NMDA-receptor independent actions of ketamine: a new chapter in a story that’s not so old , 2018, Neuropsychopharmacology.
[26] Yuko Fujita,et al. Lack of deuterium isotope effects in the antidepressant effects of (R)-ketamine in a chronic social defeat stress model , 2018, Psychopharmacology.
[27] J. Bischofberger,et al. Dendrite-targeting interneurons control synaptic NMDA-receptor activation via nonlinear α5-GABAA receptors , 2018, Nature Communications.
[28] M. Furey,et al. Neurophysiological Changes Associated with Antidepressant Response to Ketamine Not Observed in a Negative Trial of Scopolamine in Major Depressive Disorder , 2018, The international journal of neuropsychopharmacology.
[29] A. Pałucha-Poniewiera. The role of glutamatergic modulation in the mechanism of action of ketamine, a prototype rapid-acting antidepressant drug. , 2018 .
[30] Jaime E. C. Hallak,et al. Efficacy, tolerability, and safety of serotonergic psychedelics for the management of mood, anxiety, and substance-use disorders: a systematic review of systematic reviews , 2018, Expert review of clinical pharmacology.
[31] F. Vollenweider,et al. Changes in global and thalamic brain connectivity in LSD-induced altered states of consciousness are attributable to the 5-HT2A receptor , 2018, eLife.
[32] L. Ferrucci,et al. Plasma metabolomic profiling of a ketamine and placebo crossover trial of major depressive disorder and healthy control subjects , 2018, Psychopharmacology.
[33] S. Salton,et al. Role of a VGF/BDNF/TrkB Autoregulatory Feedback Loop in Rapid-Acting Antidepressant Efficacy , 2018, Journal of Molecular Neuroscience.
[34] S. Vigod,et al. Mortality risk associated with psychological distress and major depression: A population-based cohort study. , 2018, Journal of affective disorders.
[35] S. Chaki,et al. 5-HT1A receptor stimulation in the medial prefrontal cortex mediates the antidepressant effects of mGlu2/3 receptor antagonist in mice , 2018, Neuropharmacology.
[36] T. Gould,et al. Ketamine and Ketamine Metabolite Pharmacology: Insights into Therapeutic Mechanisms , 2018, Pharmacological Reviews.
[37] P. Xie,et al. Mechanisms of ketamine on mice hippocampi shown by gas chromatography–mass spectrometry-based metabolomic analysis , 2018, Neuroreport.
[38] M. Rasenick,et al. NMDAR-independent, cAMP-dependent antidepressant actions of ketamine , 2018, Molecular Psychiatry.
[39] D. E. Olson,et al. Psychedelics Promote Structural and Functional Neural Plasticity , 2018, Cell reports.
[40] J. Witkin,et al. Rapid-Acting Antidepressants. , 2018, Current pharmaceutical design.
[41] D. B. Rawlins,et al. Biopharmaceutical Characterization and Oral Efficacy of a New Rapid Acting Antidepressant Ro 25-6981. , 2018, Journal of pharmaceutical sciences.
[42] N. Galvão-Coelho,et al. Cortisol Modulation by Ayahuasca in Patients With Treatment Resistant Depression and Healthy Controls , 2018, Front. Psychiatry.
[43] K. Hashimoto,et al. (2R,6R)-Hydroxynorketamine is not essential for the antidepressant actions of (R)-ketamine in mice , 2018, Neuropsychopharmacology.
[44] J. Ramos-Quiroga,et al. Antidepressant Efficacy and Tolerability of Ketamine and Esketamine: A Critical Review , 2018, CNS Drugs.
[45] L. Giner,et al. A Review on the General Stability of Mood Disorder Diagnoses Along the Lifetime , 2018, Current Psychiatry Reports.
[46] Facundo Carrillo,et al. Natural speech algorithm applied to baseline interview data can predict which patients will respond to psilocybin for treatment-resistant depression. , 2018, Journal of affective disorders.
[47] C. Martelli,et al. Meta-analysis and review of dopamine agonists in acute episodes of mood disorder: Efficacy and safety , 2018, Journal of psychopharmacology.
[48] Hanjie Yu,et al. Vesicular glutamate transporter 1 (VGLUT1)-mediated glutamate release and membrane GluA1 activation is involved in the rapid antidepressant-like effects of scopolamine in mice , 2018, Neuropharmacology.
[49] T. Gould,et al. Mechanisms of ketamine action as an antidepressant , 2018, Molecular Psychiatry.
[50] S. Sogabe,et al. HBT1, a Novel AMPA Receptor Potentiator with Lower Agonistic Effect, Avoided Bell-Shaped Response in In Vitro BDNF Production , 2018, The Journal of Pharmacology and Experimental Therapeutics.
[51] Hailan Hu,et al. Ketamine blocks bursting in the lateral habenula to rapidly relieve depression , 2018, Nature.
[52] Hailan Hu,et al. Astroglial Kir4.1 in the lateral habenula drives neuronal bursts in depression , 2018, Nature.
[53] R. Duman,et al. Rapid-Acting Antidepressants: Mechanistic Insights and Future Directions , 2018, Current Behavioral Neuroscience Reports.
[54] Xiang Cai,et al. Protein kinase A mediates scopolamine-induced mTOR activation and an antidepressant response. , 2018, Journal of affective disorders.
[55] Xia Li,et al. Negative allosteric modulation of alpha 5-containing GABAA receptors engenders antidepressant-like effects and selectively prevents age-associated hyperactivity in tau-depositing mice , 2018, Psychopharmacology.
[56] David J. Nutt,et al. Quality of Acute Psychedelic Experience Predicts Therapeutic Efficacy of Psilocybin for Treatment-Resistant Depression , 2018, Front. Pharmacol..
[57] P. Stanton,et al. Positive N-Methyl-D-Aspartate Receptor Modulation by Rapastinel Promotes Rapid and Sustained Antidepressant-Like Effects , 2018, The international journal of neuropsychopharmacology.
[58] K. Hashimoto,et al. Mechanistic Target of Rapamycin–Independent Antidepressant Effects of (R)-Ketamine in a Social Defeat Stress Model , 2018, Biological Psychiatry.
[59] R. Duman,et al. Activity-Dependent Brain-Derived Neurotrophic Factor Release Is Required for the Rapid Antidepressant Actions of Scopolamine , 2018, Biological Psychiatry.
[60] D. Nutt,et al. Psychiatry & the psychedelic drugs. Past, present & future , 2017, Neuropharmacology.
[61] D. Nutt,et al. Increased amygdala responses to emotional faces after psilocybin for treatment-resistant depression , 2017, Neuropharmacology.
[62] Yuko Fujita,et al. Possible role of the gut microbiota–brain axis in the antidepressant effects of (R)-ketamine in a social defeat stress model , 2017, Translational Psychiatry.
[63] Yuko Fujita,et al. 5-Hydroxytryptamine-Independent Antidepressant Actions of (R)-Ketamine in a Chronic Social Defeat Stress Model , 2017, The international journal of neuropsychopharmacology.
[64] K. Hashimoto,et al. Comparison of (R)-ketamine and lanicemine on depression-like phenotype and abnormal composition of gut microbiota in a social defeat stress model , 2017, Scientific Reports.
[65] D. Nutt,et al. Psilocybin with psychological support for treatment-resistant depression: six-month follow-up , 2017, Psychopharmacology.
[66] K. Hashimoto,et al. Role of NMDA receptor GluN2D subunit in the antidepressant effects of enantiomers of ketamine. , 2017, Journal of pharmacological sciences.
[67] Evan J. Kyzar,et al. Psychedelic Drugs in Biomedicine. , 2017, Trends in pharmacological sciences.
[68] D. Nutt,et al. Psilocybin with psychological support improves emotional face recognition in treatment-resistant depression , 2017, Psychopharmacology.
[69] Kevin Murphy,et al. Psilocybin for treatment-resistant depression: fMRI-measured brain mechanisms , 2017, Scientific Reports.
[70] E. Ballard,et al. The Effect of a Single Dose of Intravenous Ketamine on Suicidal Ideation: A Systematic Review and Individual Participant Data Meta-Analysis. , 2017, The American journal of psychiatry.
[71] R. Kessler,et al. Efficacy and Safety of Selective Serotonin Reuptake Inhibitors, Serotonin-Norepinephrine Reuptake Inhibitors, and Placebo for Common Psychiatric Disorders Among Children and Adolescents: A Systematic Review and Meta-analysis , 2017, JAMA psychiatry.
[72] R. Duman,et al. Novel rapid-acting antidepressants: molecular and cellular signaling mechanisms , 2017, Neuronal signaling.
[73] K. Hashimoto,et al. (R)-Ketamine Shows Greater Potency and Longer Lasting Antidepressant Effects Than Its Metabolite (2R,6R)-Hydroxynorketamine , 2017, Biological Psychiatry.
[74] M. Popoli,et al. Acute or Chronic? A Stressful Question , 2017, Trends in Neurosciences.
[75] W. Bunney,et al. A Circadian Genomic Signature Common to Ketamine and Sleep Deprivation in the Anterior Cingulate Cortex , 2017, Biological Psychiatry.
[76] Hanjie Yu,et al. The Requirement of L-Type Voltage-Dependent Calcium Channel (L-VDCC) in the Rapid-Acting Antidepressant-Like Effects of Scopolamine in Mice , 2017, The international journal of neuropsychopharmacology.
[77] Jiang-Ping Liu,et al. Inhibition of α5 subunit‐containing GABAA receptors facilitated spinal nociceptive transmission and plasticity , 2017, European journal of pain.
[78] Matthew W. Johnson,et al. Potential Therapeutic Effects of Psilocybin , 2017, Neurotherapeutics.
[79] Zhongmin Zhang,et al. The antidepressant-like effects of biperiden may involve BDNF/TrkB signaling-mediated BICC1 expression in the hippocampus and prefrontal cortex of mice , 2017, Pharmacology Biochemistry and Behavior.
[80] S. Chaki. mGlu2/3 Receptor Antagonists as Novel Antidepressants. , 2017, Trends in pharmacological sciences.
[81] K. Martinowich,et al. BDNF at the Synapse: Why Location Matters , 2017, Molecular Psychiatry.
[82] JaneR . Taylor,et al. GLYX-13 Produces Rapid Antidepressant Responses with Key Synaptic and Behavioral Effects Distinct from Ketamine , 2017, Neuropsychopharmacology.
[83] R. Higgs,et al. Further Evaluation of Mechanisms Associated with the Antidepressantlike Signature of Scopolamine in Mice. , 2017, CNS & neurological disorders drug targets.
[84] C. Zarate,et al. New targets for rapid antidepressant action , 2017, Progress in Neurobiology.
[85] K. Hashimoto,et al. Blockade of interleukin-6 receptor in the periphery promotes rapid and sustained antidepressant actions: a possible role of gut–microbiota–brain axis , 2017, Translational Psychiatry.
[86] S. Ide,et al. Resolvin D1 and D2 Reverse Lipopolysaccharide-Induced Depression-Like Behaviors Through the mTORC1 Signaling Pathway , 2017, The international journal of neuropsychopharmacology.
[87] D. McKinzie,et al. Preclinical predictors that the orthosteric mGlu2/3 receptor antagonist LY3020371 will not engender ketamine-associated neurotoxic, motor, cognitive, subjective, or abuse-liability-related effects , 2017, Pharmacology Biochemistry and Behavior.
[88] R. Raffa,et al. What is the mechanism of Ketamine's rapid‐onset antidepressant effect? A concise overview of the surprisingly large number of possibilities , 2017, Journal of clinical pharmacy and therapeutics.
[89] K. Hashimoto,et al. Antidepressant Potential of (R)-Ketamine in Rodent Models: Comparison with (S)-Ketamine , 2017, The Journal of Pharmacology and Experimental Therapeutics.
[90] P. Ornstein,et al. Comparative Effects of LY3020371, a Potent and Selective Metabotropic Glutamate (mGlu) 2/3 Receptor Antagonist, and Ketamine, a Noncompetitive N-Methyl-d-Aspartate Receptor Antagonist in Rodents: Evidence Supporting the Use of mGlu2/3 Antagonists, for the Treatment of Depression. , 2017, The Journal of Pharmacology and Experimental Therapeutics.
[91] Jodi L. Smith,et al. In vitro pharmacological and rat pharmacokinetic characterization of LY3020371, a potent and selective mGlu2/3 receptor antagonist , 2017, Neuropharmacology.
[92] J. Dorrian,et al. Health Psychology in Australia , 2017 .
[93] S. Shetty,et al. The fibrinolytic system: A new target for treatment of depression with psychedelics. , 2017, Medical hypotheses.
[94] S. Thompson,et al. A Negative Allosteric Modulator for α5 Subunit-Containing GABA Receptors Exerts a Rapid and Persistent Antidepressant-Like Action without the Side Effects of the NMDA Receptor Antagonist Ketamine in Mice , 2017, eNeuro.
[95] D. David,et al. Ketamine treatment involves medial prefrontal cortex serotonin to induce a rapid antidepressant-like activity in BALB/cJ mice , 2017, Neuropharmacology.
[96] P. Llorca,et al. Investigational drugs in recent clinical trials for treatment-resistant depression , 2017, Expert review of neurotherapeutics.
[97] D. Araujo,et al. Rapid antidepressant effects of the psychedelic ayahuasca in treatment-resistant depression: a randomized placebo-controlled trial , 2018, Psychological Medicine.
[98] D. Lodge,et al. Selective Pharmacological Augmentation of Hippocampal Activity Produces a Sustained Antidepressant-Like Response without Abuse-Related or Psychotomimetic Effects , 2017, The international journal of neuropsychopharmacology.
[99] R. Duman,et al. Molecular and Cellular Mechanisms of Rapid-Acting Antidepressants Ketamine and Scopolamine , 2016, Current neuropharmacology.
[100] C. Nilsson,et al. Towards the Molecular Foundations of Glutamatergic-targeted Anti-depressants , 2016, Current neuropharmacology.
[101] P. Stanton,et al. The Development of Rapastinel (Formerly GLYX-13); A Rapid Acting and Long Lasting Antidepressant , 2016, Current Neuropharmacology.
[102] K. Hashimoto,et al. Adjunctive treatment of brexpiprazole with fluoxetine shows a rapid antidepressant effect in social defeat stress model: Role of BDNF-TrkB signaling , 2016, Scientific Reports.
[103] Andrzej Pilc,et al. Antidepressant-like effects of scopolamine in mice are enhanced by the group II mGlu receptor antagonist LY341495 , 2016, Neuropharmacology.
[104] R. Duman,et al. Fast-acting antidepressants rapidly stimulate ERK signaling and BDNF release in primary neuronal cultures , 2016, Neuropharmacology.
[105] Matthew W. Johnson,et al. Psilocybin produces substantial and sustained decreases in depression and anxiety in patients with life-threatening cancer: A randomized double-blind trial , 2016, Journal of psychopharmacology.
[106] B. Schmidt,et al. Rapid and sustained symptom reduction following psilocybin treatment for anxiety and depression in patients with life-threatening cancer: a randomized controlled trial , 2016, Journal of psychopharmacology.
[107] K. Hashimoto,et al. Depression-like phenotype by deletion of α7 nicotinic acetylcholine receptor: Role of BDNF-TrkB in nucleus accumbens , 2016, Scientific Reports.
[108] D. Lodge,et al. Hippocampal Perineuronal Nets Are Required for the Sustained Antidepressant Effect of Ketamine , 2016, The international journal of neuropsychopharmacology.
[109] Laura M. Geffert,et al. Rapid and sustained antidepressant properties of an NMDA antagonist/monoamine reuptake inhibitor identified via transporter-based virtual screening , 2016, Pharmacology Biochemistry and Behavior.
[110] Liang Tang,et al. Neuroprotective effects of HTR1A antagonist WAY-100635 on scopolamine-induced delirium in rats and underlying molecular mechanisms , 2016, BMC Neuroscience.
[111] T. Svensson,et al. The novel antipsychotic drug brexpiprazole, alone and in combination with escitalopram, facilitates prefrontal glutamatergic transmission via a dopamine D1 receptor-dependent mechanism , 2016, European Neuropsychopharmacology.
[112] E. Carboni,et al. Ketamine modulates catecholamine transmission in the bed nucleus of stria terminalis: The possible role of this region in the antidepressant effects of ketamine , 2016, European Neuropsychopharmacology.
[113] E. Wauson,et al. Kinase-mediated signaling cascades in mood disorders and antidepressant treatment , 2016, Journal of neurogenetics.
[114] Michael R. Johnson,et al. Adjunctive Lanicemine (AZD6765) in Patients with Major Depressive Disorder and History of Inadequate Response to Antidepressants: A Randomized, Placebo-Controlled Study , 2016, Neuropsychopharmacology.
[115] K. Hashimoto. Ketamine’s antidepressant action: beyond NMDA receptor inhibition , 2016, Expert opinion on therapeutic targets.
[116] Daniel M. Rotroff,et al. Metabolomic signatures of drug response phenotypes for ketamine and esketamine in subjects with refractory major depressive disorder: new mechanistic insights for rapid acting antidepressants , 2016, Translational psychiatry.
[117] E. Lauterbach. Treatment Resistant Depression with Loss of Antidepressant Response: Rapid-Acting Antidepressant Action of Dextromethorphan, A Possible Treatment Bridging Molecule. , 2016, Psychopharmacology bulletin.
[118] J. Petrović,et al. Molecular Mechanism and Clinical Relevance of Ketamine as Rapid‐Acting Antidepressant , 2016, Drug development research.
[119] M. Picciotto,et al. GABA interneurons mediate the rapid antidepressant-like effects of scopolamine. , 2016, The Journal of clinical investigation.
[120] James Rucker,et al. Psilocybin with psychological support for treatment-resistant depression: an open-label feasibility study. , 2016, The lancet. Psychiatry.
[121] S. Mitchell,et al. The Rapidly Acting Antidepressant Ketamine and the mGlu2/3 Receptor Antagonist LY341495 Rapidly Engage Dopaminergic Mood Circuits , 2016, The Journal of Pharmacology and Experimental Therapeutics.
[122] Xi-Ping Huang,et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites , 2016, Nature.
[123] K. Hashimoto,et al. Reduction of dopamine D2/3 receptor binding in the striatum after a single administration of esketamine, but not R-ketamine: a PET study in conscious monkeys , 2016, European Archives of Psychiatry and Clinical Neuroscience.
[124] K. Behar,et al. Transiently increased glutamate cycling in rat PFC is associated with rapid onset of antidepressant-like effects , 2016, Molecular Psychiatry.
[125] S. Chaki,et al. The Antidepressant Effects of an mGlu2/3 Receptor Antagonist and Ketamine Require AMPA Receptor Stimulation in the mPFC and Subsequent Activation of the 5-HT Neurons in the DRN , 2016, Neuropsychopharmacology.
[126] G. Aghajanian,et al. Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants , 2016, Nature Medicine.
[127] K. Hashimoto,et al. Serum n-3 polyunsaturated fatty acids and psychological distress in early pregnancy: Adjunct Study of Japan Environment and Children's Study , 2016, Translational Psychiatry.
[128] J. Riba,et al. Antidepressant Effects of a Single Dose of Ayahuasca in Patients With Recurrent Depression: A SPECT Study , 2016, Journal of clinical psychopharmacology.
[129] T. Rantamäki,et al. Antidepressant drug action — From rapid changes on network function to network rewiring , 2016, Progress in Neuro-Psychopharmacology and Biological Psychiatry.
[130] B. Dean,et al. The Role of Muscarinic Receptors in the Pathophysiology of Mood Disorders: A Potential Novel Treatment? , 2015, Current neuropharmacology.
[131] P. Stanton,et al. The long-lasting antidepressant effects of rapastinel (GLYX-13) are associated with a metaplasticity process in the medial prefrontal cortex and hippocampus , 2015, Neuroscience.
[132] Andrea Navarria,et al. Rapid antidepressant actions of scopolamine: Role of medial prefrontal cortex and M1-subtype muscarinic acetylcholine receptors , 2015, Neurobiology of Disease.
[133] R. Schwarcz,et al. The Prodrug 4-Chlorokynurenine Causes Ketamine-Like Antidepressant Effects, but Not Side Effects, by NMDA/GlycineB-Site Inhibition , 2015, The Journal of Pharmacology and Experimental Therapeutics.
[134] K. Hashimoto,et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects , 2015, Translational Psychiatry.
[135] Judith E. Hall,et al. Ketamine amplifies induced gamma frequency oscillations in the human cerebral cortex , 2015, European Neuropsychopharmacology.
[136] E. Seifritz,et al. Psilocybin-induced spiritual experiences and insightfulness are associated with synchronization of neuronal oscillations , 2015, Psychopharmacology.
[137] S. Thompson,et al. Rapid Antidepressant Action and Restoration of Excitatory Synaptic Strength After Chronic Stress by Negative Modulators of Alpha5-Containing GABAA Receptors , 2015, Neuropsychopharmacology.
[138] J. Feldon,et al. Radixin regulates synaptic GABAA receptor density and is essential for reversal learning and short-term memory , 2015, Nature Communications.
[139] K. Hashimoto,et al. Potentiation of neurite outgrowth by brexpiprazole, a novel serotonin–dopamine activity modulator: A role for serotonin 5-HT1A and 5-HT2A receptors , 2015, European Neuropsychopharmacology.
[140] Qinwen Wang,et al. PI3K/AKT/mTOR Signaling-Mediated Neuropeptide VGF in the Hippocampus of Mice Is Involved in the Rapid Onset Antidepressant-Like Effects of GLYX-13 , 2015, The international journal of neuropsychopharmacology.
[141] J. Roiser,et al. Neural correlates of change in major depressive disorder anhedonia following open-label ketamine , 2015, Journal of psychopharmacology.
[142] J. Witkin. Commentary: obstacles to the discovery of medicines for psychiatric disorders in modern times. , 2015, CNS & neurological disorders drug targets.
[143] B. Zemelman,et al. Rapid antidepressants stimulate the decoupling of GABAB receptors from GIRK/Kir3 channels through increased protein stability of 14-3-3η , 2015, Molecular Psychiatry.
[144] T. Passie,et al. LSD-assisted psychotherapy for anxiety associated with a life-threatening disease: A qualitative study of acute and sustained subjective effects , 2015, Journal of psychopharmacology.
[145] A. Grace,et al. Restoring Mood Balance in Depression: Ketamine Reverses Deficit in Dopamine-Dependent Synaptic Plasticity , 2014, Biological Psychiatry.
[146] R. E. Higgs,et al. M1 and M2 Muscarinic Receptor Subtypes Regulate Antidepressant-Like Effects of the Rapidly Acting Antidepressant Scopolamine , 2014, The Journal of Pharmacology and Experimental Therapeutics.
[147] J. Roiser,et al. Anti-anhedonic effect of ketamine and its neural correlates in treatment-resistant bipolar depression , 2014, Translational Psychiatry.
[148] Daniel Z Lieberman,et al. The utility of the combination of dextromethorphan and quinidine in the treatment of bipolar II and bipolar NOS. , 2014, Journal of affective disorders.
[149] S. Chaki,et al. Requirement of AMPA receptor stimulation for the sustained antidepressant activity of ketamine and LY341495 during the forced swim test in rats , 2014, Behavioural Brain Research.
[150] Mark D. Miller,et al. Safety, tolerability, and clinical effect of low-dose buprenorphine for treatment-resistant depression in midlife and older adults. , 2014, The Journal of clinical psychiatry.
[151] T. Passie,et al. Safety and Efficacy of Lysergic Acid Diethylamide-Assisted Psychotherapy for Anxiety Associated With Life-threatening Diseases , 2014, The Journal of nervous and mental disease.
[152] Michael K Parides,et al. Efficacy of intravenous ketamine for treatment of chronic posttraumatic stress disorder: a randomized clinical trial. , 2014, JAMA psychiatry.
[153] K. Hashimoto,et al. R (−)-ketamine shows greater potency and longer lasting antidepressant effects than S (+)-ketamine , 2014, Pharmacology Biochemistry and Behavior.
[154] Nanxin Li,et al. Scopolamine Rapidly Increases Mammalian Target of Rapamycin Complex 1 Signaling, Synaptogenesis, and Antidepressant Behavioral Responses , 2013, Biological Psychiatry.
[155] M. Quirk,et al. Lanicemine: a low-trapping NMDA channel blocker produces sustained antidepressant efficacy with minimal psychotomimetic adverse effects , 2013, Molecular Psychiatry.
[156] Carlos A. Zarate,et al. A Randomized Trial of a Low-Trapping Nonselective N-Methyl-D-Aspartate Channel Blocker in Major Depression , 2013, Biological Psychiatry.
[157] D. Nutt,et al. Anhedonia revisited: Is there a role for dopamine-targeting drugs for depression? , 2013, Journal of psychopharmacology.
[158] M. Furey,et al. Antidepressant Effects of the Muscarinic Cholinergic Receptor Antagonist Scopolamine: A Review , 2013, Biological Psychiatry.
[159] E. Kavalali,et al. The Role of Eukaryotic Elongation Factor 2 Kinase in Rapid Antidepressant Action of Ketamine , 2013, Biological Psychiatry.
[160] J. Rybakowski,et al. Possible usefulness of tianeptine in treatment-resistant depression , 2013, International journal of psychiatry in clinical practice.
[161] C. Pae,et al. Tianeptine combination for partial or non‐response to selective serotonin re‐uptake inhibitor monotherapy , 2013, Psychiatry and clinical neurosciences.
[162] P. Stanton,et al. GLYX-13, a NMDA Receptor Glycine-Site Functional Partial Agonist, Induces Antidepressant-Like Effects Without Ketamine-Like Side Effects , 2013, Neuropsychopharmacology.
[163] G. Tononi,et al. Concomitant BDNF and sleep slow wave changes indicate ketamine-induced plasticity in major depressive disorder. , 2013, The international journal of neuropsychopharmacology.
[164] H. Onoe,et al. Metabotropic glutamate 2/3 receptor antagonists improve behavioral and prefrontal dopaminergic alterations in the chronic corticosterone-induced depression model in mice , 2013, Neuropharmacology.
[165] E. Peselow,et al. Scopolamine as an Antidepressant: A Systematic Review , 2013, Clinical neuropharmacology.
[166] C. Lyketsos,et al. Mibampator (LY451395) randomized clinical trial for agitation/aggression in Alzheimer's disease , 2012, International Psychogeriatrics.
[167] J. Krystal,et al. Capturing the angel in "angel dust": twenty years of translational neuroscience studies of NMDA receptor antagonists in animals and humans. , 2012, Schizophrenia bulletin.
[168] S. Chaki,et al. Involvement of the mammalian target of rapamycin signaling in the antidepressant-like effect of group II metabotropic glutamate receptor antagonists , 2011, Neuropharmacology.
[169] J. Witkin. Commentary [the mood in the field of antidepressant drug discovery]. , 2011, CNS & neurological disorders drug targets.
[170] D. Janowsky. Serendipity Strikes Again: Scopolamine as an Antidepressant Agent in Bipolar Depressed Patients , 2011, Current psychiatry reports.
[171] J. A. Boer,et al. The cholinergic system and depression , 2011, Behavioural Brain Research.
[172] E. Kavalali,et al. NMDA Receptor Blockade at Rest Triggers Rapid Behavioural Antidepressant Responses , 2011, Nature.
[173] J. Weiss,et al. Effects of chronic antidepressant drug administration and electroconvulsive shock on activity of dopaminergic neurons in the ventral tegmentum. , 2011, The international journal of neuropsychopharmacology.
[174] C. Grob,et al. Pilot study of psilocybin treatment for anxiety in patients with advanced-stage cancer. , 2011, Archives of general psychiatry.
[175] S. Chaki,et al. AMPA receptor mediates mGlu 2/3 receptor antagonist-induced dopamine release in the rat nucleus accumbens shell , 2010, Neurochemistry International.
[176] Nanxin Li,et al. mTOR-Dependent Synapse Formation Underlies the Rapid Antidepressant Effects of NMDA Antagonists , 2010, Science.
[177] 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.
[178] G. Nowak,et al. The involvement of NMDA and AMPA receptors in the mechanism of antidepressant-like action of zinc in the forced swim test , 2010, Amino Acids.
[179] K. Hsu,et al. Activation of muscarinic acetylcholine receptors induces a nitric oxide-dependent long-term depression in rat medial prefrontal cortex. , 2010, Cerebral cortex.
[180] J. Greer,et al. Selective Antagonism of Opioid‐Induced Ventilatory Depression by an Ampakine Molecule in Humans Without Loss of Opioid Analgesia , 2010, Clinical pharmacology and therapeutics.
[181] P. Skolnick,et al. Glutamate-based antidepressants: 20 years on. , 2009, Trends in pharmacological sciences.
[182] P. Popik,et al. Zinc supplementation augments efficacy of imipramine in treatment resistant patients: a double blind, placebo-controlled study. , 2009, Journal of affective disorders.
[183] A. D. Rodrigues,et al. In Vitro and in Vivo Properties of 3-tert-Butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)-pyrazolo[1,5-d]-[1,2,4]triazine (MRK-016), a GABAA Receptor α5 Subtype-Selective Inverse Agonist , 2009, Journal of Pharmacology and Experimental Therapeutics.
[184] G. Sanacora,et al. Targeting glial physiology and glutamate cycling in the treatment of depression. , 2009, Biochemical pharmacology.
[185] B. Dean,et al. Decreased muscarinic receptor binding in the frontal cortex of bipolar disorder and major depressive disorder subjects. , 2009, Journal of affective disorders.
[186] R. Howland. The antidepressant effects of anticholinergic drugs. , 2009, Journal of psychosocial nursing and mental health services.
[187] 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.
[188] N. Scherbaum,et al. Opiate Treatment in Depression Refractory to Antidepressants and Electroconvulsive Therapy , 2008, Journal of clinical psychopharmacology.
[189] P. Skolnick. AMPA Receptors: A Target for Novel Antidepressants? , 2008, Biological Psychiatry.
[190] L. Iversen. The Monoamine Hypothesis of Depression , 2008 .
[191] B. Moghaddam,et al. NMDA Receptor Hypofunction Produces Opposite Effects on Prefrontal Cortex Interneurons and Pyramidal Neurons , 2007, The Journal of Neuroscience.
[192] D. Kupfer,et al. Acute and Longer- Term Outcomes in Depressed Outpatients Requiring One or Several Treatment Steps: A STAR*D Report , 2006 .
[193] M. Furey,et al. Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled clinical trial. , 2006, Archives of general psychiatry.
[194] 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.
[195] W. Drevets,et al. Reduced muscarinic type 2 receptor binding in subjects with bipolar disorder. , 2006, Archives of general psychiatry.
[196] R. Duman,et al. A Neurotrophic Model for Stress-Related Mood Disorders , 2006, Biological Psychiatry.
[197] E. Nisenbaum,et al. A role for AMPA receptors in mood disorders. , 2006, Biochemical pharmacology.
[198] S. Chaki,et al. A metabotropic glutamate 2/3 receptor antagonist, MGS0039, increases extracellular dopamine levels in the nucleus accumbens shell , 2006, Neuroscience Letters.
[199] S. Chaki,et al. AMPA receptor stimulation mediates the antidepressant-like effect of a group II metabotropic glutamate receptor antagonist , 2005, Brain Research.
[200] S. Chaki,et al. Neuropharmacological profiles of antagonists of group II metabotropic glutamate receptors , 2005, Neuroscience Letters.
[201] P. Érdi,et al. Modulation of septo-hippocampal θ activity by GABAA receptors: an experimental and computational approach 1 1 Supplementary data associated with this article can be found at doi:10.1016/j.neuroscience.2004.03.043. , 2004, Neuroscience.
[202] R. Traub,et al. Alpha 5 subunit-containing GABAA receptors affect the dynamic range of mouse hippocampal kainate-induced gamma frequency oscillations in vitro. , 2004, The Journal of physiology.
[203] Alan Frazer,et al. Onset and Early Behavioral Effects of Pharmacologically Different Antidepressants and Placebo in Depression , 2004, Neuropsychopharmacology.
[204] V. Novotny,et al. P.1.132 First signs of improvement with tianeptine in the treatment of depression: An analysis of a double-blind study versus fluoxetine , 2003, European Neuropsychopharmacology.
[205] K. Marthak,et al. Olanzapine and fluoxetine combination in severe or resistant depression , 2003, Indian journal of psychiatry.
[206] D. Lorrain,et al. Effects of ketamine and n-methyl-d-aspartate on glutamate and dopamine release in the rat prefrontal cortex: modulation by a group II selective metabotropic glutamate receptor agonist LY379268 , 2003, Neuroscience.
[207] P. Greengard,et al. Involvement of striatal and extrastriatal DARPP-32 in biochemical and behavioral effects of fluoxetine (Prozac) , 2002, Proceedings of the National Academy of Sciences of the United States of America.
[208] B. Moghaddam,et al. NMDA antagonist effects on striatal dopamine release: Microdialysis studies in awake monkeys , 2002, Synapse.
[209] N. Harada,et al. Cholinergic Neuronal Modulation Alters Dopamine D2Receptor Availability In Vivo by Regulating Receptor Affinity Induced by Facilitated Synaptic Dopamine Turnover: Positron Emission Tomography Studies with Microdialysis in the Conscious Monkey Brain , 2000, The Journal of Neuroscience.
[210] John H Krystal,et al. Antidepressant effects of ketamine in depressed patients , 2000, Biological Psychiatry.
[211] D. Cyril D’Souza,et al. Interactive effects of subanesthetic ketamine and haloperidol in healthy humans , 1999, Psychopharmacology.
[212] D. Brocks. Anticholinergic drugs used in Parkinson's disease: An overlooked class of drugs from a pharmacokinetic perspective. , 1999, Journal of Pharmacy & Pharmaceutical Sciences.
[213] R. Mckernan,et al. Autoradiographic localization of α5 subunit-containing GABAA receptors in rat brain , 1999, Brain Research.
[214] J. McGinty,et al. Muscarinic receptors regulate extracellular glutamate levels in the rat striatum: an in vivo microdialysis study. , 1998, The Journal of pharmacology and experimental therapeutics.
[215] P. Willner. The mesolimbic dopamine system as a target for rapid antidepressant action , 1997, International clinical psychopharmacology.
[216] J. Kelsoe,et al. No antidepressant effect of biperiden compared with placebo in depression: A double-blind 6-week clinical trial , 1995, Psychiatry Research.
[217] S. Lukas,et al. Buprenorphine treatment of refractory depression. , 1995, Journal of clinical psychopharmacology.
[218] P. Skolnick,et al. Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. , 1990, European journal of pharmacology.
[219] H. Groenewegen,et al. The distribution and compartmental organization of the cholinergic neurons in nucleus accumbens of the rat , 1989, Neuroscience.
[220] D. Nichols,et al. Differences between the mechanism of action of MDMA, MBDB, and the classic hallucinogens. Identification of a new therapeutic class: entactogens. , 1986, Journal of psychoactive drugs.
[221] James V. Hinrichs,et al. Ketamine: behavioral effects of subanesthetic doses. , 1984, Journal of clinical psychopharmacology.
[222] S. Kasper,et al. The Anticholinergic Biperiden in Depressive Disorders , 1981, Pharmacopsychiatria.
[223] A. Phillips,et al. Long-term deficits in stimulation-induced behaviors and self-stimulation after 6-hydroxydopamine administration in rats. , 1976, Behavioral biology.
[224] D. Janowsky,et al. A cholinergic-adrenergic hypothesis of mania and depression. , 1972, Lancet.
[225] C. Savage. Lysergic acid diethylamide; a clinical-psychological study. , 1952, The American journal of psychiatry.
[226] S. Chaki. mGlu2/3 receptor antagonists. , 2019, Advances in pharmacology.
[227] D. De Gregorio,et al. d-Lysergic acid diethylamide, psilocybin, and other classic hallucinogens: Mechanism of action and potential therapeutic applications in mood disorders. , 2018, Progress in brain research.
[228] K. Raab-Graham,et al. Engaging homeostatic plasticity to treat depression , 2018, Molecular Psychiatry.
[229] D. Araujo,et al. Antidepressant effects of a single dose of ayahuasca in patients with recurrent depression: a preliminary report. , 2015, Revista brasileira de psiquiatria.
[230] Toshiaki Saitoh,et al. Effect of psilocin on extracellular dopamine and serotonin levels in the mesoaccumbens and mesocortical pathway in awake rats. , 2015, Biological & pharmaceutical bulletin.
[231] J. Price,et al. Neurocircuitry of Mood Disorders , 2010, Neuropsychopharmacology.
[232] E. Nisenbaum,et al. Positive Allosteric Modulation of AMPA Receptors: A Novel Potential Antidepressant Therapy , 2010 .
[233] S. Koslow,et al. Onset of antidepressant activity: Reexamining the structure of depression and multiple actions of drugs , 1996, Depression and anxiety.
[234] W. Richards,et al. LSD-assisted psychotherapy in patients with terminal cancer. , 1973, International pharmacopsychiatry.
[235] W. Richards,et al. LSD-assisted psychotherapy with terminal cancer patients. , 1969, Current psychiatric therapies.