Elsevier

Neuropharmacology

Volume 142, November 2018, Pages 30-40
Neuropharmacology

Invited review
Some distorted thoughts about ketamine as a psychedelic and a novel hypothesis based on NMDA receptor-mediated synaptic plasticity

https://doi.org/10.1016/j.neuropharm.2018.06.008Get rights and content

Highlights

  • Ketamine, an NMDA receptor (NMDAR) antagonist, is an atypical psychedelic.

  • Its psychedelic actions may involve multiple effects.

  • STP and LTP are distinct types of plasticity induced through activation of different NMDAR subtypes.

  • Ketamine blocks STP more potently than LTP, via an action on GluN2D-containing NMDARs.

  • Inhibition of STP is a novel candidate mechanism for some effects of ketamine.

Abstract

Ketamine, a channel blocking NMDA receptor antagonist, is used off-label for its psychedelic effects, which may arise from a combination of several inter-related actions. Firstly, reductions of the contribution of NMDA receptors to afferent information from external and internal sensory inputs may distort sensations and their processing in higher brain centres. Secondly, reductions of NMDA receptor-mediated excitation of GABAergic interneurons can result in glutamatergic overactivity. Thirdly, limbic cortical disinhibition may indirectly enhance dopaminergic and serotonergic activity. Fourthly, inhibition of NMDA receptor mediated synaptic plasticity, such as short-term potentiation (STP) and long-term potentiation (LTP), could lead to distorted memories. Here, for the first time, we compared quantitatively the effects of ketamine on STP and LTP. We report that ketamine inhibits STP in a double sigmoidal fashion with low (40 nM) and high (5.6 μM) IC50 values. In contrast, ketamine inhibits LTP in a single sigmoidal manner (IC50 value ∼ 15 μM). A GluN2D-subunit preferring NMDA receptor antagonist, UBP145, has a similar pharmacological profile. We propose that the psychedelic effects of ketamine may involve the inhibition of STP and, potentially, associated forms of working memory.

This article is part of the Special Issue entitled ‘Psychedelics: New Doors, Altered Perceptions’.

Introduction

Shortly after the synthesis in the 1950s of phencyclidine (PCP) (Maddox et al., 1965) and later of ketamine (McCarthy et al., 1965), it became apparent that these two anaesthetics produced some bizarre central effects in both laboratory animals (Chen et al., 1959) and in man (Greifenstein et al., 1958; Meyer et al., 1959). The drugs produced a state of anaesthesia and analgesia with a good safety margin before lethality but poor muscle relaxation (White et al., 1982). This unusual anaesthetic state was termed ‘dissociative’ after both the disrupted electroencephalographic patterns and the sensory deprivation with detachment from the real world (Domino et al., 1965; see Domino and Luby, 2012). The central effects, following either PCP or ketamine, include dizziness, nausea, delirium, confusion, disorientation, paranoia, amnesia, dysarthria, dysphoria with agitation, unpredictable aggression and delusions of extreme strength (Allen and Young, 1978; Domino et al., 1965; Luisada, 1978; Siegel, 1978a). Both drugs were known as psychotomimetics from the outset (Luby et al., 1959), and have been used to create animal models of schizophrenia (Cadinu et al., 2017; Frohlich and Van Horn, 2014; Javitt et al., 2012) but can they also be defined as psychedelics? Psychedelics are ‘drugs (especially LSD [lysergic acid diethylamide] that produce hallucinations and apparent expansion of consciousness’ and hallucinations are experiences ‘involving the apparent perception of something not present’ (Oxford English Dictionary). There are hundreds of reports of hallucinations, visual, auditory and somatosensory, following exposure to ketamine and PCP.

For example, a recreational user of ketamine reported: ‘… when I closed my eyes a lot of information started to happen. Colors, patterns, cross-connections in sensory perception. Like sound and inner visions sort of got confused. I got deeper and deeper into this state of realization, until at one point the world disappeared. I was no longer in my body. I didn't have a body. And I reached a point at which I knew I was going to die …. .And what incredible feelings that evoked! . . . . . .I just yielded. And then I entered a space in which . . .there aren't any words ….I mean, at-one-with-the-universe, recognizing-your-godhead ….The feeling was: I was home. That's really the feeling of it. And I didn't want to go anywhere, and I didn't need to go anywhere. It was a bliss state. Of a kind I had never experienced before. I hung out there awhile, and then I came back. I didn't want to come back. I guess in the deep state it was no longer than half an hour ‘. Stafford (1977), quoted in (Siegel, 1978b) who also quotes Young et al. (1977) likening ‘this state to an LSD trip, only the ketamine tripper "feels as if he is floating in a dreamlike state while experiencing vivid visual images"’. In fact, there are numerous reports noting similarities between experiences with LSD and ketamine. But ketamine, unlike classical psychedelics such as LSD, psilocybin, mescaline and dimethyltryptamine (DMT), is not a serotonin receptor agonist and its central effects in man can be distinguished from those of LSD, as described on psychonaut websites. In humans, serotonin-based hallucinogens and dissociative anaesthetics have psychedelic features in common, as well as abuse potential, and their mode of action may be linked by direct or indirect agonism of 5-HT2 receptors (Heal et al., 2018; Sellers et al., 2017). In a standard psychometric test which scores five separate dimensions of altered states of consciousness (Dittrich, 1998), a classical psychedelic and ketamine showed similarities but clear differences (Vollenweider and Kometer, 2010). The profile following ketamine was skewed toward the dimensions of ‘disembodiment’ and ‘experience of unity’. Furthermore, in behavioural studies in laboratory animals, the effects of LSD and PCP/ketamine are easily discriminated, suggesting a quite different mode of action (Carroll, 1990; Jones and Balster, 1998; West et al., 2000).

Drug discrimination studies in rats, monkeys, pigeons, etc, have, however found many drugs that do generalize to the PCP and ketamine cues. In the early 1980s, it became apparent that PCP and ketamine provided similar cues to other arylcyclohexylamines such as tiletamine, to benzomorphan sigma opiates such as SKF10,047 and cyclazocine, to dioxalanes such as dexoxadrol and etoxadrol, to morphinans such as dextrorphan and dextromethorphan, to benz(f)isoquinolines and to propanolamines such as 2-MDP (Brady and Balster, 1981; Brady et al., 1982a, 1982b; Herrling et al., 1981; Holtzman, 1982, 1980; Mendelsohn et al., 1984; Shannon, 1982a, 1982b; 1981; Tang et al., 1984; White and Holtzman, 1982). Such a list of compounds coincides with those also shown to displace PCP binding from rat brain tissue (Hampton et al., 1982; Murray and Leid, 1984; Quirion et al., 1981; Sircar and Zukin, 1983; Vincent et al., 1979; Zukin and Zukin, 1981; Zukin, 1982; Zukin et al., 1984; Zukin and Zukin, 1979).

Several of these compounds were already known to have similar bizarre subjective effects in man including hallucinations and were generally known as psychotomimetics (see Lodge and Mercier, 2015 for many of the chemical structures). Thus, beside arylcyclohexylamines PCP and ketamine (see above), the morphinans, dextromethorphan and dextrorphan, were originally described as producing ‘toxic symptoms (dizziness, diplopia, etc)’ (Isbell and Fraser, 1953). Benzomorphans including the ‘sigma agonists’, SKF 10.047 (Keats and Telford, 1964) and cyclazocine produce ‘dose-related scores on the LSD scale’ (Haertzen, 1970 quoted by Jasinski et al., 1967) and fuzzy thinking, illusions, dysphoria and frank visual hallucinations like LSD (Freedman and Fink, 1968; Jasinski et al., 1967); such features were considered to be mediated by the sigma opiate receptor (Martin et al., 1976). The dioxalanes, dexoxadrol (Lasagna and Pearson, 1965), and etoxadrol, induced ‘dreams and/or visions that were pleasing.’ (Frederickson et al., 1976).

Further contemporary support for this psychedelic effect of such compounds comes from a cursory examination of the web sites frequented by so-called psychonauts. This reveals that drugs such as ketamine, dextromethorphan, 2-MDP and compounds related to them are still frequently being used by humans. To avoid legal issues associated with ketamine use, new drugs for example, methoxetamine and arylethylamines, such as ephenidine, have come on to the market (Kang et al., 2016; Morris and Wallach, 2014; Wallach et al., 2016). During the last two weeks of January 2018, for example on one website, there were 35 reports of experiences with these and similar drugs (https://erowid.org/experiences/exp.cgi?New).

Section snippets

Sites of action of ketamine and PCP

So do these compounds have a mechanism of action in common with classical psychedelics such as LSD, mescaline, psilocybin and dimethyltryptamine, which are all serotonergic ligands? It is generally believed that their agonist effect on 5-HT2 receptors, particularly 5-HT2A receptors, is the basis for their psychedelic effects (Nichols, 2004; Vollenweider and Kometer, 2010). Activation of 5-HT2A receptors also increases glutamate release and firing of pyramidal neurones in the cerebral cortex by

Effects of ketamine on central information processing

So the question arises as to why does NMDA receptor antagonism induce psychedelic effects? Here we discuss three potential mechanisms based on NMDAR antagonism that individually or collectively could contribute to these effects.-

  • 1.

    Direct and/or indirect activation of dopaminergic pathways has been proposed to underlie the psychotomimetic effects of ketamine and related compounds.

  • 2.

    A reduction in synaptic transmission through afferent pathways in which NMDA receptors play an important role will

Conclusions

Ketamine is a relatively selective NMDA receptor antagonist. Because NMDA receptors are known to contribute to synaptic events throughout the brain and spinal cord, ketamine will distort afferent information, processing in the cerebral cortex and output pathways. Specifically, because of their biophysical properties, NMDA receptor heteromers containing the GluN2D subtype are particularly sensitive to ketamine. This relatively high potency of ketamine at GluN2D subunits and their importance in

Conflicts of interest

The authors have no competing interests to declare.

Acknowledgments

Supported by The Royal Society (RSG∖R1∖180384), the MRC (MR/K023098/1) and CIHR.

References (158)

  • R.E. Garey et al.

    The effects of phencyclidine on the uptake of 3H-catecholamines by rat striatal and hypothalamic synaptosomes

    Life Sci.

    (1976)
  • R.Y. Hampton et al.

    Stereospecific binding of 3H-phencyclidine in brain membranes

    Life Sci.

    (1982)
  • J.D. Jentsch et al.

    The neuropsychopharmacology of phencyclidine: from NMDA receptor hypofunction to the dopamine hypothesis of schizophrenia

    Neuropsychopharmacology

    (1999)
  • H.E. Jones et al.

    Muscimol-like discriminative stimulus effects of GABA agonists in rats

    Pharmacol. Biochem. Behav.

    (1998)
  • R.S. Jones et al.

    Basket-like interneurones in layer II of the entorhinal cortex exhibit a powerful NMDA-mediated synaptic excitation

    Neurosci. Lett.

    (1993)
  • S.E. Kotermanski et al.

    Comparison of behavioral effects of the NMDA receptor channel blockers memantine and ketamine in rats

    Pharmacol. Biochem. Behav.

    (2013)
  • D.A. Lewis

    Inhibitory neurons in human cortical circuits: substrate for cognitive dysfunction in schizophrenia

    Curr. Opin. Neurobiol.

    (2014)
  • D. Lodge et al.

    Effects of ketamine and three other anaesthetics on spinal reflexes and inhibitions in the cat

    Br. J. Anaesth.

    (1984)
  • D. Lodge et al.

    Effect of ketamine on amino acid-evoked release of acetylcholine from rat cerebral cortex in vitro

    Neurosci. Lett.

    (1985)
  • R.C. Malenka

    Postsynaptic factors control the duration of synaptic enhancement in area CA1 of the hippocampus

    Neuron

    (1991)
  • G.J. Marek et al.

    5-HT2A receptor or alpha1-adrenoceptor activation induces excitatory postsynaptic currents in layer V pyramidal cells of the medial prefrontal cortex

    Eur. J. Pharmacol.

    (1999)
  • D. Martin et al.

    Ketamine acts as a non-competitive N-methyl-D-aspartate antagonist on frog spinal cord in vitro

    Neuropharmacology

    (1985)
  • L.G. Mendelsohn et al.

    Phencyclidine receptors in rat brain cortex

    Biochem. Pharmacol.

    (1984)
  • R. Moaddel et al.

    Sub-anesthetic concentrations of (R,S)-ketamine metabolites inhibit acetylcholine-evoked currents in α7 nicotinic acetylcholine receptors

    Eur. J. Pharmacol.

    (2013)
  • H. Monyer et al.

    Developmental and regional expression in the rat brain and functional properties of four NMDA receptors

    Neuron

    (1994)
  • K.W. Muir

    Glutamate-based therapeutic approaches: clinical trials with NMDA antagonists

    Curr. Opin. Pharmacol.

    (2006)
  • E.D. Adrian

    The Basis of Sensation: the Action of the Sense Organs

    (1928)
  • M. Agetsuma et al.

    Parvalbumin-Positive interneurons regulate neuronal ensembles in visual cortex

    Cereb. Cortex

    (2017)
  • R.M. Allen et al.

    Phencyclidine-induced psychosis

    Am. J. Psychiatry

    (1978)
  • N.A. Anis et al.

    The dissociative anaesthetics, ketamine and phencyclidine, selectively reduce excitation of central mammalian neurones by N-methyl-aspartate

    Br. J. Pharmacol.

    (1983)
  • T.V. Bliss et al.

    A synaptic model of memory: long-term potentiation in the hippocampus

    Nature

    (1993)
  • K.T. Brady et al.

    Stereoisomers of N-allylnormetazocine: phencyclidine-like behavioral effects in squirrel monkeys and rats

    Science

    (1982)
  • K.T. Brady et al.

    Discriminative stimulus and reinforcing properties of etoxadrol and dexoxadrol in monkeys

    J. Pharmacol. Exp. Ther.

    (1982)
  • R.G. Browne et al.

    Stereoselective antagonism of phencyclidine's discriminative properties by adenosine receptor agonists

    Science

    (1982)
  • D. Cadinu et al.

    NMDA receptor antagonist rodent models for cognition in schizophrenia and identification of novel drug treatments, an update

    Neuropharmacology

    (2017)
  • M.E. Carroll

    PCP and hallucinogens

    Adv. Alcohol Subst. Abuse

    (1990)
  • G. Chen et al.

    The pharmacology of 1-(1-phenylcyclohexyl) piperidine-HCl

    J. Pharmacol. Exp. Ther.

    (1959)
  • K. Chergui et al.

    Tonic activation of NMDA receptors causes spontaneous burst discharge of rat midbrain dopamine neurons in vivo

    Eur. J. Neurosci.

    (1993)
  • G.L. Collingridge et al.

    Excitatory amino acids in synaptic transmission in the Schaffer collateral-commissural pathway of the rat hippocampus

    J. Physiol. (Lond.)

    (1983)
  • J.T. Coyle et al.

    Beyond the dopamine receptor: novel therapeutic targets for treating schizophrenia

    Dialogues Clin. Neurosci.

    (2010)
  • S.M. Davis et al.

    Selfotel in acute ischemic stroke : possible neurotoxic effects of an NMDA antagonist

    Stroke

    (2000)
  • D. Debanne et al.

    Heterogeneity of synaptic plasticity at unitary CA3-CA1 and CA3-CA3 connections in rat hippocampal slice cultures

    J. Neurosci.

    (1999)
  • A. Dittrich

    The standardized psychometric assessment of altered states of consciousness (ASCs) in humans

    Pharmacopsychiatry

    (1998)
  • E.F. Domino et al.

    Pharmacologic effects of CI-581, a new dissociative anesthetic, in man

    Clin. Pharmacol. Ther.

    (1965)
  • E.F. Domino et al.

    Phencyclidine/schizophrenia: one view toward the past, the other to the future

    Schizophr. Bull.

    (2012)
  • S.M. Dravid et al.

    Subunit-specific mechanisms and proton sensitivity of NMDA receptor channel block

    J. Physiol. (Lond.)

    (2007)
  • S.M. Dudek et al.

    Homosynaptic long-term depression in area CA1 of hippocampus and effects of N-methyl-D-aspartate receptor blockade

    Proc. Natl. Acad. Sci. U.S.A.

    (1992)
  • N.B. Farber et al.

    Serotonergic agents that activate 5HT2A receptors prevent NMDA antagonist neurotoxicity

    Neuropsychopharmacology

    (1998)
  • E.L. Frederickson et al.

    Clinical investigation of a new intravenous anesthetic–etoxadrol hydrochloride (CL-1848; U-37862A)

    Anesth. Analg.

    (1976)
  • A.M. Freedman et al.

    Basic concepts and use of cyclazocine in the treatment of narcotic addiction

    Br. J. Addiction Alcohol Other Drugs

    (1968)
  • Cited by (0)

    1

    DL and AV are joint senior authors.

    View full text