Effects of the GluN2B-selective antagonist Ro 63-1908 on acquisition and expression of methamphetamine conditioned place preference in male and female rats

https://doi.org/10.1016/j.drugalcdep.2021.108785Get rights and content

Highlights

  • Ro 63-1908 blocks acquisition of methamphetamine CPP in male rats.

  • Ro 63-1908 attenuates acquisition of methamphetamine CPP in female rats.

  • Ro 63-1908 does not block expression of methamphetamine-induced place preference.

  • Ro 63-1908 (1.0 mg/kg) enhances the locomotor-stimulant effects of methamphetamine.

  • Ro 63-1908 was neither rewarding nor aversive when administered alone.

Abstract

Background

Methamphetamine abuse has increased significantly in recent years. Currently, there are no FDA-approved pharmacotherapies for the treatment of methamphetamine use disorder. The goal of the current study was to determine if the N-methyl-d-aspartate (NMDA) GluN2B-selective antagonist Ro 63-1908 can block the conditioned rewarding effects of methamphetamine as assessed in conditioned place preference (CPP).

Methods

Two main experiments were conducted. In the first experiment, male (n = 24) and female (n = 24) rats received either vehicle or Ro 63-1908 (1.0–10.0 mg/kg) 30 min prior to the posttest to determine if blocking the GluN2B subunit attenuates expression of methamphetamine CPP. In the second experiment, male (n = 18) and female (n = 18) rats received either vehicle or Ro 63-1908 (1.0 or 3.0 mg/kg) 30 min prior to each conditioning session to determine if blocking the GluN2B subunit attenuates acquisition of methamphetamine CPP.

Results

Ro 63-1908 (3.0 mg/kg) blocked acquisition of methamphetamine CPP in male rats, but only attenuated CPP in female rats. Ro 63-1908 did not alter expression of CPP in either sex. Increasing the dose of Ro 63-1908 (10.0 mg/kg) failed to block acquisition of CPP in an additional group of female rats (n = 6). A control experiment showed that Ro 63-1908 (3.0 mg/kg) did not produce CPP or conditioned place aversion in male rats (n = 6) or in female rats (n = 6).

Conclusions

The results of this study show that Ro 63-1908 is able to decrease the conditioned rewarding effects of methamphetamine.

Introduction

Methamphetamine is a psychostimulant drug that has been prescribed to treat attention-deficit/hyperactivity disorder (ADHD) (as Desoxyn) that is reinforcing in several species (e.g., Balster and Schuster, 1973; Harrod et al., 2001; Hart et al., 2001; Munzar et al., 1999) and has high abuse potential (see Courtney and Ray, 2014). Because there are no FDA-approved pharmacotherapies for treating methamphetamine use disorder, finding a treatment option has been of considerable interest in recent years. Several pharmacological treatments have been used to reduce methamphetamine use, including agonist replacement therapy (i.e., amphetamine and methylphenidate) (Galloway et al., 2011; Miles et al., 2013; Rezaei et al., 2015; Rush et al., 2011; but see Pike et al., 2014 for mixed results), antidepressant medications such as bupropion (Ahmadi et al., 2019; Anderson et al., 2015; Elkashef et al., 2008; Heinzerling et al., 2014; Newton et al., 2006; Shoptaw et al., 2008, but see Stoops et al., 2015) and mirtazapine (Colfax et al., 2011), and the mu opioid receptor antagonist naltrexone (Anggadiredja et al., 2004; but see Coffin et al., 2018; Stoops et al., 2015). Even though several drug classes have been shown to reduce methamphetamine use or decrease cravings in individual studies, results of a recent meta-analysis revealed that most drugs reviewed did not provide statistically significant reductions in methamphetamine use (Chen et al., 2019). The results of this meta-analysis highlight the need to test novel molecular targets for the treatment of methamphetamine use disorder.

Although methamphetamine’s primary mechanisms of action are to reverse the vesicular monoamine transporter (VMAT2) (Peter et al., 1994) and monoamine transporters, such as the dopamine transporter (DAT) (Eschleman et al., 1994), as well as to inhibit monoamine oxidase (MAO) (Suzuki et al., 1980), there is evidence that methamphetamine directly interacts with the glutamatergic system. Glutamate is the major excitatory neurotransmitter in the mammalian brain and binds to several types of metabotropic receptors (mGluRs) and ionotropic receptors (iGluRs), including N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (see Ozawa et al., 1998 for a review). In humans, acute administration of methamphetamine increases glutamate/glutamine levels in dorsal anterior cingulate cortex in women, but not in men (White et al., 2018), but decreased levels of glutamate/glutamine are observed in recently abstinent men and women (Ernst and Chang, 2008). Methamphetamine significantly increases evoked excitatory currents within prefrontal cortex (PFC) in male rats and mice (Han et al., 2012; Lominac et al., 2016; Mishra et al., 2017; but see Pena-Bravo et al., 2019) and in female rats (Pena-Bravo et al., 2019). In male rodents, methamphetamine increases extracellular levels of glutamate in striatum (Bustamante et al., 2002; Nash and Yamamoto, 1992) and medial prefrontal cortex (mPFC) (Han et al., 2012), decreases extracellular glutamate levels in dorsal hippocampus (Han et al., 2012), alters NMDA receptor subunit and vesicular glutamate transporter (VGLUT2) expression in striatum (Furlong et al., 2018; Simões et al., 2008), and increases GluN1 and GluN2B subunit expression in PFC (Mishra et al., 2017; Simões et al., 2008) and in hippocampus (Li et al., 2014; Simões et al., 2007). Additionally, repeated methamphetamine administration decreases NMDA receptor binding in PFC and hippocampus (Lee et al., 2011). Overall, this evidence suggests that targeting the glutamatergic system may provide novel treatment options for methamphetamine use disorder.

To this end, the NMDA receptor channel blocker MK-801 decreases methamphetamine conditioned place preference (CPP) (Kim and Jang, 1997). Considering MK-801 is a psychotomimetic that can cause Olney’s lesions (Olney et al., 1989), using this drug as a therapeutic for methamphetamine use disorder is unrealistic. Although all NMDA receptors are composed of two NR1 subunits, these receptors are also composed of two NR2 subunits that can be subdivided into GluN2A-D. The GluN2B subunit has received considerable attention, as ifenprodil, a GluN2B antagonist, is able to attenuate the rewarding effects of opiates (Ma et al., 2006, 2011; Suzuki et al., 1999), prevent reinstatement of nicotine-seeking behavior (Gipson et al., 2013), and block the acquisition of methamphetamine CPP (Kurokawa et al., 2011).

Although Kurokawa et al. (2011) have previously shown that ifenprodil can block the conditioned rewarding effects of methamphetamine, they only tested the effects of ifenprodil on the acquisition of CPP. That is, they administered ifenprodil prior to each methamphetamine injection. In order to determine if a drug is effective in treating a substance use disorder, it is important to test the effects of the potential pharmacotherapy after animals have already acquired the association between an environmental context and the drug of abuse. This is known as the expression of CPP. Ifenprodil is able to block the expression of morphine CPP (e.g., Ma et al., 2006), but research has not determined if GluN2B-selective antagonists can block the expression of methamphetamine CPP. Additionally, Kurokawa et al. (2011) only tested male mice in their study. Given that a large number of preclinical studies have used male subjects exclusively (see second paragraph of Introduction), the goal of the current study was to determine if the highly selective GluN2B-selective antagonist Ro 63-1908 is able to attenuate the expression and/or the acquisition of methamphetamine CPP in males and in females. We selected Ro 63-1908 because it has higher affinity for the GluN2B subunit compared to ifenprodil (Gill et al., 2002; Higgins et al., 2005).

Section snippets

Subjects

A total of 48 male (241.354 ± 1.271 g/64.143 ± 0.451 days old at the beginning of testing) and 48 female (178.417 ± 1.234 g/52.146 ± 1.085 days old at the beginning of testing) Sprague Dawley rats (Envigo, Indianapolis, IN) were used in the present experiments. Rats were housed individually immediately upon delivery to the laboratory. Rats were housed in a temperature- and humidity-controlled colony room that was maintained on a light–dark cycle in which lights were on from 7:00 a.m. to 7:00

Time spent in each compartment during pretest

Table 1 shows the time spent in each compartment during the pretest for the expression, the acquisition, and the control experiments. For all experiments, there was a main effect of compartment, all F’s ≥ 10.436, all p’s ≤ .001, all f’s ≥ 0.906. For each experiment, Tukey’s post hoc tests showed that the time spent in the gray compartment was lower than the time spent in the white compartment and in the black compartment. In the expression experiment, Tukey’s post hoc tests also showed that the

Discussion

In the current experiment, we intentionally used a dose of methamphetamine that has been shown to produce CPP in males and in females (Hensleigh and Pritchard, 2014; Risca et al., 2020; Schindler et al., 2002) as we wanted to determine if Ro 63-1908 could attenuate the acquisition and/or expression of methamphetamine CPP to a similar extent across each sex. We found that Ro 63-1908 (3.0 mg/kg) blocks the acquisition of methamphetamine CPP in male rats and attenuates, but not completely blocks,

Role of funding source

None of the funding sources were involved in the study design, analysis, interpretation, or writing of the current manuscript.

Contributors

Justin Yates conceptualized the experiments, conducted behavioral testing, analyzed the data, wrote the manuscript, and revised the manuscript. Hunter Campbell and Lauren Hawley conceptualized the experiments, conducted behavioral testing, and wrote the manuscript. Matthew Horchar conducted behavioral testing and revised the manuscript. Joy Kappesser and Makayla Wright conducted behavioral testing.

Declaration of Competing Interest

The authors report no declarations of interest.

Acknowledgements

The current study was supported by NIH grant R15DA047610 and NIGMS grant P20GM103436. The study was also supported by a Northern Kentucky University Faculty Project Grant and a Northern Kentucky University College of Arts and Sciences Professional Development Award.

References (84)

  • G.A. Higgins et al.

    Enhanced attention and impulsive action following NMDA receptor GluN2B-selective antagonist pretreatment

    Behav. Brain Res.

    (2016)
  • E. Jerlhag et al.

    Ghrelin receptor antagonism attenuates nicotine-induced locomotor stimulation, accumbal dopamine release and conditioned place preference in mice

    Drug Alcohol Depend.

    (2011)
  • M. Jiang et al.

    Inhibiting effects of rhynchophylline on zebrafish methamphetamine dependence are associated with amelioration of neurotransmitters content and down-regulation of TH and NR2B expression

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2016)
  • L. Jing et al.

    Involvement of dopamine D2 receptor in a single methamphetamine-induced behavioral sensitization in C57BL/6J mice

    Neurosci. Lett.

    (2018)
  • J.-H. Kao et al.

    NR2B subunit of NMDA receptor at nucleus accumbens is involved in morphine rewarding effect by siRNA study

    Drug Alcohol Depend.

    (2011)
  • H.S. Kim et al.

    MK-801 inhibits methamphetamine-induced conditioned place preference and behavioral sensitization to apomorphine in mice

    Brain Res. Bull.

    (1997)
  • H.S. Kim et al.

    Inhibition by MK-801 of cocaine-induced sensitization, conditioned place preference, and dopamine-receptor supersensitivity in mice

    Brain Res. Bull.

    (1996)
  • D.H. Kim et al.

    Sauchinone blocks methamphetamine-induced hyperlocomotion and place preference in mice

    Phytomedicine

    (2013)
  • K.W. Lee et al.

    Methamphetamine-sensitized mice are accompanied by memory impairment and reduction of N-methyl-D-aspartate receptor ligand binding in the prefrontal cortex and hippocampus

    Neuroscience

    (2011)
  • J. Li et al.

    Effect of rhynchophylline on conditioned place preference on expression of NR2B in methamphetamine-dependent mice

    Biochem. Biophys. Res. Commun.

    (2014)
  • L. Li et al.

    Ifenprodil attenuates the acquisition and expression of methamphetamine-induced behavioral sensitization and activation of Ras-ERK1/2 cascade in caudate putamen

    Neuroscience

    (2016)
  • J.M. Lima-Ojeda 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

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2013)
  • Y.Y. Ma et al.

    The role of NR2B containing NMDA receptor in place preference conditioned with morphine and natural reinforcers in rats

    Exp. Neurol.

    (2006)
  • P. Mareš et al.

    Different effects of two N-methyl-D-aspartate receptor antagonists on seizures, spontaneous behavior, and motor performance in immature rats

    Epilepsy Behav.

    (2009)
  • P. Mikolajczak et al.

    Effects of acamprosate and some polyamine site ligands of NMDA receptor on short-term memory in rats

    Eur. J. Pharmacol.

    (2002)
  • J.F. Nash et al.

    Methamphetamine neurotoxicity and striatal glutamate release: comparison to 3,4-methylendioxymethamphetamine

    Brain Res.

    (1992)
  • S. Ozawa et al.

    Glutamate receptors in the mammalian central nervous system

    Prog. Neurobiol.

    (1998)
  • D. Peter et al.

    The chomaffin granule and synaptic vesicle amine transporters differ in substrate recognition and sensitivity to inhibitors

    J. Biol. Chem.

    (1994)
  • H.I. Risca et al.

    Conditioned place preference following concurrent treatment with 3,4-methylenedioxypyrovalerone (MDPV) and methamphetamine in male and female Sprague-Dawley rats

    Pharmacol. Biochem. Behav.

    (2020)
  • K.R. Rodvelt et al.

    The sigma receptor agonist SA4503 both attenuates and enhances the effects of methamphetamine

    Drug Alcohol Depend.

    (2011)
  • C.W. Schindler et al.

    Gender differences in the behavioral effects of methamphetamine

    Eur. J. Pharmacol.

    (2002)
  • E.-J. Shin et al.

    Attenuation of cocaine-induced conditioned place preference by Polygala tenuifolia root extract

    Life Sci.

    (2004)
  • S. Shoptaw et al.

    Randomized, placebo-controlled trial of bupropion for the treatment of methamphetamine dependence

    Drug Alcohol Depend.

    (2008)
  • P.F. Simões et al.

    Methamphetamine induces alterations on hippocampal NMDA and AMPA receptor subunit levels and impairs spatial working memory

    Neuroscience

    (2007)
  • W.W. Stoops et al.

    Naltrexone and bupropion, alone or combined, do not alter the reinforcing effects of intranasal methamphetamine

    Pharmacol. Biochem. Behav.

    (2015)
  • O. Suzuki et al.

    Inhibition of monoamine oxidase by d-methamphetamine

    Biochem. Pharmacol.

    (1980)
  • Y. Wang et al.

    Prenatal chronic mild stress induces depression-like behavior and sex-specific changes in regional glutamate receptor expression patterns in adult rats

    Neuroscience

    (2015)
  • J.U. Adams et al.

    Differential effects of dopamine antagonists on locomotor activity, conditioned activity and conditioned place preference induced by cocaine in rats

    Behav. Pharmacol.

    (2001)
  • J. Ahmadi et al.

    A randomized clinical trial on the effects of bupropion and buprenorphine on the reduction of methamphetamine craving

    Trials

    (2019)
  • D.A. Baker et al.

    Effects of intraaccumbens administration of SCH-23390 on cocaine-induced locomotion and conditioned place preference

    Synapse

    (1998)
  • D. Bustamante et al.

    Effect of single and repeated methamphetamine treatment on neurotransmitter release in substantia nigra and neostriatum of the rat

    J. Neurochem.

    (2002)
  • B. Chen et al.

    Pharmacotherapy for methamphetamine/amphetamine use disorder – a systematic review and meta-analysis

    Addiction

    (2019)
  • Cited by (8)

    • Sex differences in methamphetamine use disorder perused from pre-clinical and clinical studies: Potential therapeutic impacts

      2022, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      Interestingly, Bernheim et al. (2017) have suggested that the attenuating effects of oxytocin on METH-seeking behavior may be dependent on regulation of presynaptic glutamatergic signaling because reinstatement of lever pressing to METH-associated cues was observed after administration of LY341495, metabotropic glutamate receptor (mGluR2/3) antagonist in both male and female rats. The glutamate ionotropic receptor NMDA type subunit 2B (GluN2B) selective antagonist, Ro 63–1908, also completely blocked the conditioned rewarding effects of METH in male, but not female rats (Yates et al., 2021). Whereas administration of Ro 25–6981, another GluN2B selective antagonist, did not exhibit any sex differences in reducing METH seeking behavior (Westbrook and Gulley, 2020).

    • Effects of NMDA receptor antagonists on behavioral economic indices of cocaine self-administration

      2022, Drug and Alcohol Dependence
      Citation Excerpt :

      Recent studies have implicated GluN2B-containing NMDA receptors in behavioral phenotypes associated with vulnerability to SUDs, such as risk taking and impulsivity (Yates et al., 2018, 2021b). Furthermore, the GluN2B subunit is an important mediator of methamphetamine reward (Kurokawa et al., 2011; Yates et al., 2021a), as well as reinstatement of heroin- and nicotine-seeking behavior (Shen et al., 2011; Gipson et al., 2013). The primary goal of the current experiment was to determine if the GluN2B subunit-selective antagonist Ro 63–1908 decreases the direct reinforcing effects of cocaine.

    • Blocking serotonin 2A (5-HT<inf>2A</inf>) receptors attenuates the acquisition of methamphetamine-induced conditioned place preference in adult female rats

      2021, Behavioural Brain Research
      Citation Excerpt :

      Moreover, whether methamphetamine pharmacokinetic sex differences manifest into behavioral differences in methamphetamine-induced CPP is yet to be determined. However, preliminary work using the 1 mg/kg dose of methamphetamine has not revealed robust sex differences in methamphetamine-induced CPP [34–36]. Although direct comparisons of methamphetamine-induced CPP between the present study with females and our previous study with males is not feasible [20], the magnitude of CPP appears lower in female than in male rats using the 1 mg/kg dose of methamphetamine.

    View all citing articles on Scopus
    View full text