Fast-acting antidepressant activity of ketamine: highlights on brain serotonin, glutamate, and GABA neurotransmission in preclinical studies

https://doi.org/10.1016/j.pharmthera.2019.02.017Get rights and content

Abstract

Ketamine, a non-competitive antagonist of N-methyl-D-aspartate (NMDA) receptor, displays a fast antidepressant activity in treatment-resistant depression and in rodent models of anxiety/depression. A large body of evidence concerning the cellular and molecular mechanisms underlying its fast antidepressant-like activity comes from animal studies. Although structural remodeling of frontocortical/hippocampal neurons has been proposed as critical, the role of excitatory/inhibitory neurotransmitters in this behavioral effect is unclear. Neurochemical and behavioral changes are maintained 24h after ketamine administration, well beyond its plasma elimination half-life. Thus, ketamine is believed to initiate a cascade of cellular mechanisms supporting its fast antidepressant-like activity. To date, the underlying mechanism involves glutamate release, then downstream activation of AMPA receptors, which trigger mammalian target of rapamycin (mTOR)-dependent structural plasticity via brain-derived neurotrophic factor (BDNF) and protein neo-synthesis in the medial prefrontal cortex (mPFC), a brain region strongly involved in ketamine therapeutic effects. However, these mPFC effects are not restricted to glutamatergic pyramidal cells, but extend to other neurotransmitters (GABA, serotonin), glial cells, and brain circuits (mPFC/dorsal raphe nucleus-DRN). It could be also mediated by one or several ketamine metabolites (e.g., (2R,6R)-HNK). The present review focuses on evidence for mPFC neurotransmission abnormalities in major depressive disorder (MDD) and their potential impact on neural circuits (mPFC/DRN). We will integrate these considerations with results from recent preclinical studies showing that ketamine, at antidepressant-relevant doses, induces neuronal adaptations that involve the glutamate-excitatory/GABA-inhibitory balance. Our analyses will help direct future studies to further elucidate the mechanism of action of fast-acting antidepressant drugs, and to inform development of novel, more efficacious therapeutics.

Introduction

Major depressive disorder (MDD) is a serious, debilitating, life-shortening illness and a leading cause of disability worldwide. According to the World Health Organization, about 350 million people are affected by depression, with a higher risk for females than males (WHO, 2012). MDD is also predicted to be the second leading cause of disease burden in 2020 (Murray & Lopez, 1996).

The current most widely prescribed drugs for MDD treatment, i.e., selective serotonin reuptake inhibitors (SSRIs) and serotonin–norepinephrine reuptake inhibitors (SNRIs), display serious limitations such as the delay between drug administration and antidepressant efficacy. Divergent roles of serotonin (5-HT) autoreceptors and heteroreceptors in modulating responses to antidepressant drugs explain, at least in part, this long delay of action (Nautiyal & Hen, 2017). In addition, an up to 30% of resistance and non-response rate has made these current treatments less reliable (Mrazek, Hornberger, Altar, & Degtiar, 2014). More precisely, the response rates to SSRI antidepressant drugs are about one-third after the initial prescription and up to two-thirds after multiple drug trials.

Ketamine, a non-competitive antagonist of the N-methyl-D-aspartate subtype of excitatory amino acid receptor (NMDA-R), is a dissociative anesthetic (Krystal et al., 1994). Clinical studies have demonstrated that it displays antidepressant efficacy in treatment-resistant depression (TRD) (Berman et al., 2000; Zarate Jr. et al., 2006). Different methods have been used for staging TRD (Ruhe, van Rooijen, Spijker, Peeters, & Schene, 2012). Each staging method has its own criteria for treatment duration, classes and number of antidepressant trials, as well as severity of depression. Although there is a lack of consensus regarding these criteria of TRD, failure to respond to more than two classes of antidepressant drugs with adequate dosage and for an adequate duration is typically defined as TRD (McIntyre et al., 2014). More than fifty percent of depressed patients meet this definition (Thomas et al., 2013), among them, those taking monoaminergic antidepressant drugs of the serotonergic/noradrenergic class. However, low response rates to the first antidepressant prescription suggest that there are other strategies/mechanisms of action that may help patients with TRD (Duman & Aghajanian, 2012; Kim & Na, 2016). Ketamine could be one of them.

Twelve randomized clinical trials confirmed that ketamine is the only NMDA-R antagonist to date consistently demonstrating antidepressant efficacy in TRD. Its antidepressant effects following an intravenous infusion are both rapid and robust, even though it has a short-lived action. In most cases, most clinical evidence demonstrates significant effects up to one week with subsequent decreases in response at later time points (see meta-analysis reviews: (Caddy, Giaroli, White, Shergill, & Tracy, 2014; Fond et al., 2014; Newport, et al., 2015; Romeo, Choucha, Fossati, & Rotge, 2015; Xu, Hackett, et al., 2016).

Although downstream neuronal adaptations might be involved in its antidepressant activity (Browne & Lucki, 2013; Sanacora, Zarate, Krystal, & Manji, 2008), the exact mechanisms underlying ketamine's effects remain incompletely resolved. Analyzing in detail brain regions and circuitry involved in its mechanism of action is crucial. It has been postulated that ketamine exerts its therapeutic effect with limited direct interactions with 5-HT systems. Our recent studies performed in BALB/cJ mice suggested evidence of activation of 5-HT neurotransmission in the medial prefrontal cortex (mPFC) (Pham et al., 2017; Pham et al., 2018).

The present review will focus on preclinical data regarding cellular and molecular mechanisms of ketamine underlying its antidepressant-like activity rather than clinical data on ketamine's benefits on TRD. Indeed, a lot of clinical reviews have already been published, though many of them have used a small number of patients with TRD (see meta-analyses: (Caddy et al., 2015; Kokkinou, Ashok, & Howes, 2018; Papadimitropoulou, Vossen, Karabis, Donatti, & Kubitz, 2017)). In addition, some clinical and biological predictors of ketamine’s response are often identical to other therapeutics used in MDD; thus, it is critical to identify more specific clinical biomarkers of this response (Romeo, Choucha, Fossati, & Rotge, 2017).

Preclinical studies have begun to elucidate a working mechanism underlying the rapid antidepressant-like activity of ketamine in animal models/tests of anxiety/depression. Specifically, an initial glutamate burst and activity-dependent synapse formation in the mPFC has been shown (Duman & Aghajanian, 2012; Duman, Aghajanian, Sanacora, & Krystal, 2016; Li et al., 2010). Additionally, the involvement of the balance between excitatory (glutamate, 5-HT) and inhibitory (γ-aminobutyric acid - GABA) neurotransmission within the glutamate mPFC/5-HT dorsal raphe nucleus (DRN) circuitry in rodent studies will be addressed here. Since ketamine is comprised of a mixture of two optical isomers: (S)-ketamine and (R)-ketamine, several active metabolites such as (2R,6R)-hydroxynorketamine (HNK) may also potentially make an important contribution to its antidepressant-like activity (Can et al., 2016). Beyond NMDA-R activation, a possible direct activation of α-amino-3-hyroxy-5-methyl-4-isoxazolepropionic acid receptor subtype (AMPA-R) by (2R,6R)-HNK, the main brain active metabolite (Zanos et al., 2016), is also discussed. Lastly, we also discuss the role of several factors, including species, route and dose of administration, and timing of measures (30 min, 24 h or 7 days prior to testing), which may explain some of the inconsistencies described in the literature.

Section snippets

Pharmacodynamics and pharmacokinetics of ketamine

Ketamine is described as a powerful NMDA-R antagonist: in vitro, EC50 = 760 nM, in vivo, ED50 = 4.4 mg/kg in rodents’ cortex or hippocampus (Lord et al., 2013; F. Murray et al., 2000). Under physiological conditions and in the presence of extracellular Mg2+, the NMDA channel is closed, due to a decreased permeability to Ca2+ and an inhibition of currents mediated by NMDA-R. This process should theoretically inhibit the ability of ketamine to bind to its phencyclidine-like site (Fig. 1).

Quick view of effects of an acute ketamine administration

A sub-anesthetic dose of ketamine impairs prefrontal cortex (PFC) function in rats, which produces symptoms similar to schizophrenia in human. The pioneer experimental work of Moghaddam and colleagues (Moghaddam, Adams, Verma, & Daly, 1997) demonstrated that a single sub-anesthetic dose of ketamine activated glutamatergic neurotransmission in the PFC. Ketamine induced a rapid increase (starting at 40 min after an i.p. injection of 10, 20 or 30 mg/kg, and lasting for 100 min for the 30 mg/kg

Serotonin (5-HT)

This part of the review focuses on the knowledge that links ketamine to the brain serotonergic system. However, we have chosen not to focus on the molecular proteins/events that specifically occur in 5-HT neurons such as tryptophan hydroxylase 2 - TPH2, Pet1 and DRN signaling cascade behind 5-HT receptors, because there are many recent publications detailing these points (i.e., du Jardin et al., 2016; Hamon & Blier, 2013).

Serotonergic neurons of the mammalian brain comprise the most extensive

Conclusions

In this review, we have analyzed current data on ketamine antidepressant-like activity that involved the glutamatergic, GABAergic and serotonergic neurotransmissions.

The current indirect cortical disinhibition hypothesis regarding the cascade of cellular and molecular events leading to a fast antidepressant-like activity of an acute ketamine dose can be summarized as follows: ketamine binds to NMDA-R located on GABAergic interneurons, and induces a selective blockade of inhibitory GABA

Conflict of interest

The authors declare no conflict of interest.

Acknowledgements

We would like to thank Denis J. David and Jean-Philippe Guilloux for helpful discussions on the manuscript. We are very grateful to Josephine McGowan from Columbia University (NewYork, USA) for her careful reading of the manuscript.

References (389)

  • A. Barbon et al.

    Chronic antidepressant treatments induce a time-dependent up-regulation of AMPA receptor subunit protein levels

    Neurochemistry International

    (2011)
  • A. Barbon et al.

    Regulation of editing and expression of glutamate alpha-amino-propionic-acid (AMPA)/kainate receptors by antidepressant drugs

    Biological Psychiatry

    (2006)
  • P. Baumbarger et al.

    LY392098, a novel AMPA receptor potentiator: electrophysiological studies in prefrontal cortical neurons

    Neuropharmacology

    (2001)
  • R.M. Berman et al.

    Antidepressant effects of ketamine in depressed patients

    Biological Psychiatry

    (2000)
  • Z. Bhagwagar et al.

    Reduction in occipital cortex gamma-aminobutyric acid concentrations in medication-free recovered unipolar depressed and bipolar subjects

    Biological Psychiatry

    (2007)
  • C. Björkholm et al.

    Ketamine-like effects of a combination of olanzapine and fluoxetine on AMPA and NMDA receptor-mediated transmission in the medial prefrontal cortex of the rat

    Eur Neuropsychopharmacol

    (2015)
  • R.A. Brachman et al.

    Ketamine as a Prophylactic Against Stress-Induced Depressive-like Behavior

    Biological Psychiatry

    (2016)
  • A. Brancato et al.

    Sub-chronic variable stress induces sex-specific effects on glutamatergic synapses in the nucleus accumbens

    Neuroscience

    (2017)
  • M. Chatterjee et al.

    Neurochemical and molecular characterization of ketamine-induced experimental psychosis model in mice

    Neuropharmacology

    (2012)
  • A. Cipriani et al.

    Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: a systematic review and network meta-analysis

    Lancet

    (2018)
  • B.R. Cornwell et al.

    Synaptic potentiation is critical for rapid antidepressant response to ketamine in treatment-resistant major depression

    Biological Psychiatry

    (2012)
  • J.F. Cryan et al.

    Don't worry ‘B’ happy!: a role for GABA(B) receptors in anxiety and depression

    Trends in Pharmacological Sciences

    (2005)
  • J.F. Cryan et al.

    Assessing antidepressant activity in rodents: recent developments and future needs

    Trends in Pharmacological Sciences

    (2002)
  • E. Dale et al.

    The multimodal antidepressant vortioxetine may facilitate pyramidal cell firing by inhibition of 5-HT3 receptor expressing interneurons: An in vitro study in rat hippocampus slices

    Brain Research

    (2018 Jun 15)
  • D.J. David et al.

    Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression

    Neuron

    (2009)
  • X.F. Ding et al.

    Involvement of the glutamate/glutamine cycle and glutamate transporter GLT-1 in antidepressant-like effects of Xiao Yao san on chronically stressed mice

    BMC Complementary and Alternative Medicine

    (2017)
  • R.J. Douglas et al.

    Mapping the matrix: the ways of neocortex

    Neuron

    (2007)
  • M. Farahmandfar et al.

    Recovery from ketamine-induced amnesia by blockade of GABA-A receptor in the medial prefrontal cortex of mice

    Neuroscience

    (2017)
  • C. Fee et al.

    Somatostatin-Positive Gamma-Aminobutyric Acid Interneuron Deficits in Depression: Cortical Microcircuit and Therapeutic Perspectives

    Biological Psychiatry

    (2017)
  • D. Felice et al.

    Blockade of the GABA(B) receptor increases neurogenesis in the ventral but not dorsal adult hippocampus: relevance to antidepressant action

    Neuropharmacology

    (2012)
  • E.A. Forster et al.

    A pharmacological profile of the selective silent 5-HT1A receptor antagonist, WAY-100635

    European Journal of Pharmacology

    (1995)
  • F. Freudenberg et al.

    The role of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in depression: central mediators of pathophysiology and antidepressant activity?

    Neuroscience and Biobehavioral Reviews

    (2015)
  • J. Abbasi

    Ketamine minus the trip: New hope for treatment-resistant depression

    JAMA

    (2017)
  • C.G. Abdallah et al.

    Ketamine and rapid-acting antidepressants: a window into a new neurobiology for mood disorder therapeutics

    Annual Review of Medicine

    (2015)
  • C.A. Altamura et al.

    Plasma and platelet excitatory amino acids in psychiatric disorders

    The American Journal of Psychiatry

    (1993)
  • M. Amargos-Bosch et al.

    Clozapine and olanzapine, but not haloperidol, suppress serotonin efflux in the medial prefrontal cortex elicited by phencyclidine and ketamine

    The International Journal of Neuropsychopharmacology

    (2006)
  • B.K. Andrasfalvy et al.

    Impaired regulation of synaptic strength in hippocampal neurons from GluR1-deficient mice

    The Journal of Physiology

    (2003)
  • F. Artigas et al.

    Pindolol augmentation of antidepressant response

    Current Drug Targets

    (2006)
  • A.E. Autry et al.

    NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses

    Nature

    (2011)
  • R. Balazs

    Trophic effect of glutamate

    Current Topics in Medicinal Chemistry

    (2006)
  • M. Banasr et al.

    Glial pathology in an animal model of depression: reversal of stress-induced cellular, metabolic and behavioral deficits by the glutamate-modulating drug riluzole

    Molecular Psychiatry

    (2010)
  • J. Barnes et al.

    Genetic Contributions of Inflammation to Depression

    Neuropsychopharmacology

    (2017)
  • M.M. Behrens et al.

    Ketamine-induced loss of phenotype of fast-spiking interneurons is mediated by NADPH-oxidase

    Science

    (2007)
  • M.A. Belenky et al.

    Subcellular distribution of 5-HT(1B) and 5-HT(7) receptors in the mouse suprachiasmatic nucleus

    The Journal of Comparative Neurology

    (2001)
  • E. Beurel et al.

    Ketamine-induced inhibition of glycogen synthase kinase-3 contributes to the augmentation of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor signaling

    Bipolar Disorders

    (2016)
  • Z. Bhagwagar et al.

    Increased brain GABA concentrations following acute administration of a selective serotonin reuptake inhibitor

    The American Journal of Psychiatry

    (2004)
  • P. Blier et al.

    Long-term 5-HT reuptake blockade, but not monoamine oxidase inhibition, decreases the function of terminal 5-HT autoreceptors: an electrophysiological study in the rat brain

    Naunyn-Schmiedeberg's Archives of Pharmacology

    (1988)
  • G. Bonanno et al.

    Chronic antidepressants reduce depolarization-evoked glutamate release and protein interactions favoring formation of SNARE complex in hippocampus

    The Journal of Neuroscience

    (2005)
  • N.G. Bowery et al.

    International Union of Pharmacology. XXXIII. Mammalian gamma-aminobutyric acid(B) receptors: structure and function

    Pharmacological Reviews

    (2002)
  • P.A. Boyer et al.

    Chronic administration of imipramine and citalopram alters the expression of NMDA receptor subunit mRNAs in mouse brain. A quantitative in situ hybridization study

    Journal of Molecular Neuroscience

    (1998)
  • Cited by (118)

    View all citing articles on Scopus
    View full text