Elsevier

Biological Psychiatry

Volume 83, Issue 11, 1 June 2018, Pages 947-954
Biological Psychiatry

Review
Metabotropic Glutamate Receptors 2 and 3 as Targets for Treating Nicotine Addiction

https://doi.org/10.1016/j.biopsych.2017.11.021Get rights and content

Abstract

Tobacco smoking, driven by the addictive properties of nicotine, continues to be a worldwide health problem. Based on the well-established role of glutamatergic neurotransmission in drug addiction, novel medication development strategies seek to halt nicotine consumption and prevent relapse to tobacco smoking by modulating glutamate transmission. The presynaptic inhibitory metabotropic glutamate receptors 2 and 3 (mGluR2/3) are key autoreceptors on glutamatergic terminals that maintain glutamate homeostasis. Accumulating evidence suggests the critical role of mGluR2/3 in different aspects of nicotine addiction, including acquisition and maintenance of nicotine taking, nicotine withdrawal, and persistent nicotine seeking even after prolonged abstinence. The involvement of mGluR2/3 in other neuropsychiatric conditions, such as anxiety, depression, schizophrenia, Alzheimer’s disease, Parkinson’s disease, and pain, provides convincing evidence suggesting that mGluR2/3 may provide an effective therapeutic approach for comorbidity of smoking and these conditions. This focused review article highlights that mGluR2/3 provide a promising target in the search for smoking cessation medication with novel mechanisms of actions that differ from those of currently U.S. Food and Drug Administration–approved pharmacotherapies.

Section snippets

Neurobiology of mGluR2/3

Protein and messenger RNA expression studies reveal mGluR2/3 to be distributed in brain regions and circuits associated with neuropsychiatric disorders, including drug addiction. High density of mGluR2/3 has been identified in brain areas related to processing of reward, motivated behaviors, learning, memory, and emotion, including the nucleus accumbens (NAc), dorsal striatum, hippocampus, amygdala, prefrontal cortex (PFC), thalamus, and olfactory bulb 24, 25, 26. Although the distributions of

mGluR2/3 and Nicotine Reward

Nicotine addiction is characterized by the persistence of compulsive nicotine taking, the emergence of aversive withdrawal symptoms on abstinence from smoking (e.g., anhedonia, anxiety, craving, and irritability), and relapse to tobacco smoking even after periods of abstinence (37). Tobacco smoking or nicotine administration produces mild euphoria, increased arousal, relaxation, and decreased fatigue in humans. The rewarding effects and conditioned reinforcement of nicotine drive the

mGluR2/3 and Nicotine Withdrawal

In humans, nicotine withdrawal is associated with negative affective symptoms, such as anxiety, depression, and irritability (37). In contrast to the somatic signs that are primarily peripherally mediated, the centrally mediated negative affective withdrawal signs are hypothesized to critically contribute to the persistence of nicotine taking and relapse to smoking after abstinence 61, 64, 65, 66, 67, 68. Hence, amelioration of these affective signs of early nicotine withdrawal may prevent

mGluR2/3 and Reinstatement of Nicotine Seeking

A high rate of relapse to smoking is a major hurdle to long-term quitting. Maintenance of tobacco smoking is largely driven by conditioned motivational effects of environmental stimuli (e.g., the sight and smell of a lit cigarette or contexts within which smoking occurs) previously associated with smoking 93, 94. These conditioned cues also play an important role in relapse to smoking in humans 95, 96, 97. Smokers report strong urges to smoke after being exposed to specific environments in

mGluR2/3 and Smoking-Associated Psychiatric Disorders

As the primary excitatory neurotransmitter in the brain, glutamate is implicated in a wide range of brain functions. In addition to drug addiction, imbalances in glutamate neurotransmission have been implicated in the pathophysiology of different neurological and psychiatric conditions, such as anxiety disorders, major depression, schizophrenia, and chronic pain. As the key regulators of glutamate homeostasis, mGluR2/3 appear to be critically involved in these conditions and have been

Medication Development Targeting mGluR2/3

Owing to the high level of conservation within the extracellular orthosteric binding site of mGluR2 and mGluR3 (117), all the well-validated compounds currently commercially available bind with similar affinity to both receptors, making it impossible to distinguish the function of each receptor with these pharmacological tools. In recent years, selective mGluR2 PAMs that act on less well-conserved allosteric receptor sites within transmembrane spanning domains have been synthesized 40, 41, 118.

Acknowledgments and Disclosures

This work was supported by the National Institute on Drug Abuse Grant Nos. R56DA011946 (to AM) and R21DA040195 (to XL), Tobacco-Related Disease Research Program, University of California, Grant Nos. 20KT-0046 (to XL) and 24RT-0034 (to XL), AstraZeneca (AM), and National Institute on Drug Abuse and Department of Veterans Affairs Cooperative Studies Program Grant Nos. 032 and 1033 (to RA).

AJC and XL report no biomedical financial interests or potential conflicts of interest. RA provides

References (120)

  • Z. Mateo et al.

    Group II metabotropic glutamate receptors inhibit glutamate release at thalamocortical synapses in the developing somatosensory cortex

    Neuroscience

    (2007)
  • R.G. Greenslade et al.

    Selective action of (-)-2-oxa-4-aminobicyclo[3.1.0]hexane-4,6-dicarboxylate (LY379268), a group II metabotropic glutamate receptor agonist, on basal and phencyclidine-induced dopamine release in the nucleus accumbens shell

    Neuropharmacology

    (2004)
  • J. Karasawa et al.

    A metabotropic glutamate 2/3 receptor antagonist, MGS0039, increases extracellular dopamine levels in the nucleus accumbens shell

    Neurosci Lett

    (2006)
  • Z. Justinova et al.

    The novel metabotropic glutamate receptor 2 positive allosteric modulator, AZD8529, decreases nicotine self-administration and relapse in squirrel monkeys

    Biol Psychiatry

    (2015)
  • W.A. Corrigall et al.

    Self-administered nicotine activates the mesolimbic dopamine system through the ventral tegmental area

    Brain Res

    (1994)
  • J.A. Dani et al.

    Synaptic plasticity and nicotine addiction

    Neuron

    (2001)
  • H.D. Mansvelder et al.

    Long-term potentiation of excitatory inputs to brain reward areas by nicotine

    Neuron

    (2000)
  • P.J. Kenny et al.

    The ups and downs of addiction: Role of metabotropic glutamate receptors

    Trends Pharmacol Sci

    (2004)
  • H.D. Mansvelder et al.

    Synaptic mechanisms underlie nicotine-induced excitability of brain reward areas

    Neuron

    (2002)
  • G.F. Koob

    Neurobiological substrates for the dark side of compulsivity in addiction

    Neuropharmacology

    (2009)
  • A.H. Weinberger et al.

    Nicotine withdrawal in U.S. smokers with current mood, anxiety, alcohol use, and substance use disorders

    Drug Alcohol Depend

    (2010)
  • A.M. Abrantes et al.

    The role of negative affect in risk for early lapse among low distress tolerance smokers

    Addict Behav

    (2008)
  • S. Shiffman et al.

    Prediction of lapse from associations between smoking and situational antecedents assessed by ecological momentary assessment

    Drug Alcohol Depend

    (2007)
  • A.K. Stoker et al.

    Affective and somatic aspects of spontaneous and precipitated nicotine withdrawal in C57BL/6J and BALB/cByJ mice

    Neuropharmacology

    (2008)
  • J.M. Hogle et al.

    Nicotine withdrawal increases threat-induced anxiety but not fear: Neuroadaptation in human addiction

    Biol Psychiatry

    (2010)
  • M.E. Liechti et al.

    Interactive effects of the mGlu5 receptor antagonist MPEP and the mGlu2/3 receptor antagonist LY341495 on nicotine self-administration and reward deficits associated with nicotine withdrawal in rats

    Eur J Pharmacol

    (2007)
  • M.E. Liechti et al.

    Metabotropic glutamate 2/3 receptor activation induced reward deficits but did not aggravate brain reward deficits associated with spontaneous nicotine withdrawal in rats

    Biochem Pharmacol

    (2007)
  • L.A. Knackstedt et al.

    The role of cystine-glutamate exchange in nicotine dependence in rats and humans

    Biol Psychiatry

    (2009)
  • G. Bedi et al.

    Incubation of cue-induced cigarette craving during abstinence in human smokers

    Biol Psychiatry

    (2011)
  • D. Caprioli et al.

    Effect of the novel positive allosteric modulator of metabotropic glutamate receptor 2 AZD8529 on incubation of methamphetamine craving after prolonged voluntary abstinence in a rat model

    Biol Psychiatry

    (2015)
  • J.E. Rose et al.

    Role of nicotine dose and sensory cues in the regulation of smoke intake

    Pharmacol Biochem Behav

    (1993)
  • C.P. O’Brien et al.

    Myths about the treatment of addiction

    Lancet

    (1996)
  • A.R. Caggiula et al.

    Cue dependency of nicotine self-administration and smoking

    Pharmacol Biochem Behav

    (2001)
  • C.D. Mathers et al.

    Projections of global mortality and burden of disease from 2002 to 2030

    PLoS Med

    (2006)
  • J.J. McNeil et al.

    Smoking cessation-recent advances

    Cardiovasc Drugs Ther

    (2010)
  • A. Markou

    Review. Neurobiology of nicotine dependence

    Philos Trans R Soc Lond B Biol Sci

    (2008)
  • S.R. Laviolette et al.

    The neurobiology of nicotine addiction: Bridging the gap from molecules to behaviour

    Nat Rev Neurosci

    (2004)
  • P.J. Kenny et al.

    NMDA receptors regulate nicotine-enhanced brain reward function and intravenous nicotine self-administration: Role of the ventral tegmental area and central nucleus of the amygdala

    Neuropsychopharmacology

    (2009)
  • P.J. Kenny et al.

    Metabotropic glutamate 5 receptor antagonist MPEP decreased nicotine and cocaine self-administration but not nicotine and cocaine-induced facilitation of brain reward function in rats

    Ann N Y Acad Sci

    (2003)
  • N.E. Paterson et al.

    The mGluR5 antagonist MPEP decreased nicotine self-administration in rats and mice

    Psychopharmacology

    (2003)
  • A.S. Adewale et al.

    Pharmacological stimulation of group II metabotropic glutamate receptors reduces cocaine self-administration and cocaine-induced reinstatement of drug seeking in squirrel monkeys

    J Pharmacol Exp Ther

    (2006)
  • M.A. Baptista et al.

    Preferential effects of the metabotropic glutamate 2/3 receptor agonist LY379268 on conditioned reinstatement versus primary reinforcement: Comparison between cocaine and a potent conventional reinforcer

    J Neurosci

    (2004)
  • M.E. Liechti et al.

    Metabotropic glutamate 2/3 receptors in the ventral tegmental area and the nucleus accumbens shell are involved in behaviors relating to nicotine dependence

    J Neurosci

    (2007)
  • A. Bisaga et al.

    Therapeutic potential of NMDA receptor antagonists in the treatment of alcohol and substance use disorders

    Expert Opin Invest Drugs

    (2000)
  • P.J. Conn et al.

    Promise of mGluR2/3 activators in psychiatry

    Neuropsychopharmacology

    (2009)
  • M.F. Olive

    Metabotropic glutamate receptor ligands as potential therapeutics for addiction

    Curr Drug Abuse Rev

    (2009)
  • P.M. Lea et al.

    Metabotropic glutamate receptor subtype 5 antagonists MPEP and MTEP

    CNS Drug Rev

    (2006)
  • M.E. Liechti et al.

    Role of the glutamatergic system in nicotine dependence: Implications for the discovery and development of new pharmacological smoking cessation therapies

    CNS Drugs

    (2008)
  • A. Markou

    The role of metabotropic glutamate receptors in drug reward, motivation and dependence

    Drug News Perspect

    (2007)
  • F. Ferraguti et al.

    Metabotropic glutamate receptors

    Cell Tissue Res

    (2006)
  • Cited by (21)

    • The relationship between exposure to general anesthetic agents and the risk of developing an impulse control disorder

      2021, Pharmacological Research
      Citation Excerpt :

      mGluRs are primarily presynaptic inhibitory receptors suppressing Glu excitability. One study has demonstrated that activation of mGluRs 2/3 in the terminals of glutamatergic neurons inhibited glutamate release [85], resulting in impulsive nicotine-seeking behavior [86]. Sevoflurane anesthesia impaired the inhibitory function of mGluR through upregulating small conductance calcium-activated potassium type 2 channels [87].

    • Adolescent neurodevelopment and substance use: Receptor expression and behavioral consequences

      2020, Pharmacology and Therapeutics
      Citation Excerpt :

      The long-term effects of adolescent nicotine exposure on mGluR down-regulation in the PFC, VTA, and NAc, and the resulting behavioral and cognitive deficits are also of interest (Counotte et al., 2011). The mGluR2/3 subtype is most often implicated in nicotine addiction and self-administration (Counotte et al., 2011; Cross, Anthenelli, & Li, 2018; Liechti, Lhuillier, Kaupmann, & Markou, 2007). Lower adulthood mGluR2 expression in the rat PFC following adolescent, but not adult, nicotine exposure is also thought to underlie lasting attentional deficits (Counotte et al., 2011).

    • Allosteric modulation of metabotropic glutamate receptors in alcohol use disorder: Insights from preclinical investigations

      2020, Advances in Pharmacology
      Citation Excerpt :

      A list of allosteric modulators used in preclinical investigations of AUD and other substance use disorders (SUDs) can be found in Table 1. Although it is outside the scope of this chapter, there is extensive literature detailing preclinical investigations of mGlu receptor orthosteric and allosteric ligands on reward, consumption, and seeking of other psychoactive drugs, and we refer readers to several recent reviews that address these topics (Barnes, Sheffler, Semenova, Cosford, & Bespalov, 2018; Caprioli, Justinova, Venniro, & Shaham, 2018; Cross, Anthenelli, & Li, 2018; Joffe, Centanni, Jaramillo, Winder, & Conn, 2018; Johnson & Lovinger, 2016; Li & Markou, 2015; Scofield et al., 2016). A variety of organisms and behavioral paradigms have been used to examine the actions of acute and chronic ethanol that are thought to contribute to drinking.

    View all citing articles on Scopus
    View full text