Elsevier

Drug and Alcohol Dependence

Volume 146, 1 January 2015, Pages 7-16
Drug and Alcohol Dependence

Differential effects of endocannabinoid catabolic inhibitors on morphine withdrawal in mice

https://doi.org/10.1016/j.drugalcdep.2014.11.015Get rights and content

Highlights

  • Morphine or clonidine reduces the development of conditioned place aversions of withdrawal in morphine-dependent mice.

  • THC and endocannabinoid degradative enzyme inhibitors reduce somatic signs of morphine withdrawal, but not the development of conditioned place aversions of withdrawal.

  • Endocannabinoid degradative enzymes inhibitors do not elicit rewarding effects on their own, as assessed in the conditioned place preference paradigm.

Abstract

Background

Inhibition of endocannabinoid catabolic enzymes fatty acid amide hydrolase (FAAH) and/or monoacylglycerol lipase (MAGL) reduces somatic morphine withdrawal signs, but its effects on aversive aspects of withdrawal are unknown. The present study investigated whether Δ9-tetrahydrocannabinol (THC), the MAGL inhibitor JZL184, the FAAH inhibitor PF-3845, or the dual FAAH/MAGL inhibitor SA-57 would reduce acquisition of morphine withdrawal-induced conditioned place avoidance (CPA) and jumping.

Methods

Mice were implanted with placebo or 75 mg morphine pellets, 48 h later injected with naloxone or saline and placed in the conditioning apparatus, and assessed for CPA at 72 h. Subjects were also observed for jumping behavior following naloxone challenge.

Results

Naloxone (0.056 mg/kg) produced robust CPA in morphine-pelleted, but not placebo-pelleted, mice. Morphine pretreatment prevented the occurrence of withdrawal CPA and withdrawal jumping, while clonidine (an α2 adrenergic receptor agonist) only blocked withdrawal CPA. THC, JZL184, and SA-57 significantly reduced the percentage of mice that jumped during the conditioning session, but did not affect acquisition of withdrawal CPA. PF-3845 did not reduce morphine withdrawal CPA or jumping. Finally, neither THC nor the endocannabinoid catabolic enzyme inhibitors in non-dependent mice elicited a conditioned place preference or aversion.

Conclusions

These findings suggest that inhibiting endocannabinoid catabolic enzymes reduces somatic morphine withdrawal signs, but not aversive aspects as inferred in the CPA paradigm. The observation that non-dependent mice administered inhibitors of endocannabinoid degradation did not display place preferences is consistent with the idea that that endocannabinoid catabolic enzymes might be targeted therapeutically, with reduced risk of abuse.

Introduction

Opioid abuse and dependence continue to present a serious threat to public health (Johnston et al., 2010). Fear of withdrawal symptoms that include diarrhea, emesis, body aches, anxiety, dysphoria (Farrell, 1994, Gossop, 1988, Jasinski, 1981, Wesson and Ling, 2003) are thought to contribute to the maintenance of drug-taking in opioid dependent individuals. Likewise, continued opioid use alleviates the withdrawal state, thus serving as a negative reinforcer (Koob and Le Moal, 2005). Current available treatments for opioid dependence, such as methadone and buprenorphine, possess their own abuse liability (Cicero and Inciardi, 2005) and are not fully effective at alleviating withdrawal (Dyer et al., 1999, Kuhlman et al., 1998). Thus, new pharmacotherapies that lack abuse potential are needed to alleviate opioid withdrawal.

Extracts from cannabis and the primary constituent of marijuana, Δ9-tetrahydrocannabinol (THC), have long been known to ameliorate somatic morphine withdrawal signs (Birch, 1889, Hine et al., 1975). THC produces the bulk of its pharmacological effects through two known G-protein coupled receptors, cannabinoid type-1 (CB1; Matsuda et al., 1990) and type-2 (CB2; Munro et al., 1993). These receptors, as well as the endogenous cannabinoids (endocannabinoids) 2-arachidonoylglycerol (2-AG; Mechoulam et al., 1995, Sugiura et al., 1995) and N-arachidonoylethanolamine (anandamide, AEA; Devane et al., 1992) comprise the endogenous cannabinoid system. These endocannabinoids are rapidly degraded by the respective enzymes fatty acid amide hydrolase (FAAH; Cravatt et al., 2001) and monoacylglycerol lipase (MAGL; Dinh et al., 2002). Selective inhibitors of these endocannabinoid degradative enzymes reduce somatic signs of opioid withdrawal (e.g., jumping, paw fluttering, head/body shaking, weight loss, diarrhea; Ramesh et al., 2013, Ramesh et al., 2011). However, it is unknown whether the anti-withdrawal effects extend to the affective components of morphine withdrawal.

Opioid-dependent individuals undergoing withdrawal experience aversive subjective effects, a process that is modeled in the Pavlovian conditioned place avoidance (CPA) paradigm. In this assay morphine-dependent rats (Gracy et al., 2001, Hand et al., 1988, Parker and Rennie, 1992, Schnur et al., 1992, Stinus et al., 2000, Stinus et al., 1990, Watanabe et al., 2003) or mice (Broseta et al., 2005, Maldonado et al., 2003, Olson et al., 2006, Sato et al., 2005, Shoblock and Maidment, 2005) undergo conditioning trials in which naloxone precipitates an aversive interoceptive stimulus that is paired with a distinct chamber. Following subsequent placement into the test apparatus, the subjects spend less time in the conditioning chamber than in the control chamber (i.e., CPA). In this assay, lower doses of naloxone produce CPA than those doses necessary to elicit somatic withdrawal signs (Caillé et al., 1999, Frenois et al., 2002). Furthermore, the α2 adrenergic agonist clonidine, which is known to reduce opioid withdrawal in humans (Gold et al., 1978, Gossop, 1988), attenuates opioid withdrawal CPA (Kosten, 1994, Schulteis et al., 1998a). Given the colocalization of CB1 and mu opioid receptors in the locus coeruleus (Scavone et al., 2010), periaqueductal gray (Wilson-Poe et al., 2012) and nucleus accumbens (Pickel et al., 2004), cannabinoid receptors are advantageously positioned to compensate for the hyperactivity in neurons that are key to the expression of both somatic and aversive aspects of opioid withdrawal (Frenois et al., 2002, Lane-Ladd et al., 1997, Nestler and Tallman, 1988, Stinus et al., 1990, Widnell et al., 1994).

The purpose of the present study was to test whether stimulation of CB1 receptors via administration of THC or inhibition of endocannabinoid catabolic enzymes would prevent the acquisition of naloxone-precipitated morphine withdrawal CPA and withdrawal-related jumping behavior in mice. To this end, the MAGL inhibitor JZL184 (Long et al., 2009a) or FAAH inhibitor PF-3845 (Ahn et al., 2009) was administered at the time of conditioning. In addition, we evaluated whether the dual FAAH/MAGL inhibitor SA-57 (Niphakis et al., 2012) would prevent morphine withdrawal CPA. Of note, combined inhibition of these two major endocannabinoid hydrolytic enzymes elicits enhanced cannabimimetic activity compared with single enzyme inhibition (Long et al., 2009b, Seillier et al., 2014, Wise et al., 2012). Moreover, each of these inhibitors reduces naloxone-precipitated and spontaneous somatic withdrawal signs in morphine-dependent mice (Ramesh et al., 2013, Ramesh et al., 2011). Finally, each inhibitor was tested in non-dependent mice to ascertain whether the compounds elicited reward-related or aversive effects in the place conditioning assay.

Section snippets

Subjects

Male ICR mice (6–8 weeks old; Harlan, Indianapolis, IN) with a body mass of 27–32 g were used for all experiments. Mice were group-housed (four per cage) on a 12/12 light/dark cycle (lights on at 0600 h) and given food and water ad libitum. All animal protocols were approved by the Virginia Commonwealth University Institutional Animal Care and Use Committee, were in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals (Institute of Laboratory Animal

Naloxone-precipitated morphine withdrawal produces conditioned place avoidance behavior at a dose that does not elicit this effect in non-dependent mice

Initial experiments were designed to determine the optimal dose of naloxone that produced CPA in morphine dependent mice, but not in placebo-pelleted mice. The dose–response relationship of naloxone (0.03, 0.056, and 0.1 mg/kg) was examined in morphine- and placebo-pelleted mice. Naloxone produced a significant CPA in morphine-pelleted mice [F(3,74) = 4.3; p < 0.01; Fig. 1A] as well as in placebo-pelleted animals [F(3,80) = 4.3; p < 0.01; Fig. 1B]. Post hoc analyses revealed that morphine-pelleted mice

Discussion

The experiments in the present study tested whether inhibition of endocannabinoid catabolic enzymes or THC administration would reduce aversive aspects of morphine withdrawal. This hypothesis was tested using a Pavlovian conditioning procedure in which morphine-dependent mice display a CPA to a compartment associated with naloxone-precipitated withdrawal. Neither THC nor the endocannabinoid catabolic inhibitors significantly reduced morphine withdrawal CPA, though each of these compounds, with

Author disclosures

Role of funding source

Financial support was provided by the National Institutes of Health grants [T32DA007027, R01DA032933, P01DA009789, and P01DA01725] and a Toni Rosenberg Fellowship. The funding source had no involvement in the research reported in this manuscript or the writing of the manuscript.

Contributors

T.F. Gamage conducted the bulk of the studies, data analyses, and writing of the manuscript. B.M. Ignatowska-Jankowska conducted a subset of the experiments as well as contributed to the

References (83)

  • T.A. Kosten

    Clonidine attenuates conditioned aversion produced by naloxone-precipitated opiate withdrawal

    Eur. J. Pharmacol.

    (1994)
  • J. Liu et al.

    Brain reward deficits accompany naloxone-precipitated withdrawal from acute opioid dependence

    Pharmacol. Biochem. Behav.

    (2004)
  • C. Maldonado et al.

    Memantine presents different effects from MK-801 in motivational and physical signs of morphine withdrawal

    Behav. Brain Res.

    (2003)
  • R. Mechoulam et al.

    Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors

    Biochem. Pharmacol.

    (1995)
  • A. Mendiguren et al.

    CB1 cannabinoid receptors inhibit the glutamatergic component of KCl-evoked excitation of locus coeruleus neurons in rat brain slices

    Neuropharmacology

    (2007)
  • L.A. Parker et al.

    Naltrexone-induced aversions: assessment by place conditioning, taste reactivity, and taste avoidance paradigms

    Pharmacol. Biochem. Behav.

    (1992)
  • V. Pickel et al.

    Compartment-specific localization of cannabinoid 1 (CB1) and [mu]-opioid receptors in rat nucleus accumbens

    Neuroscience

    (2004)
  • M. Sato et al.

    Barium potentiates the conditioned aversion to, but not the somatic signs of, morphine withdrawal in mice

    Eur. J. Pharmacol.

    (2005)
  • J. Scavone et al.

    Cannabinoid and opioid interactions: implications for opiate dependence and withdrawal

    Neuroscience

    (2013)
  • J.L. Scavone et al.

    Characterization of cannabinoid-1 receptors in the locus coeruleus: relationship with mu-opioid receptors

    Brain Res.

    (2010)
  • G.J. Schaefer et al.

    Morphine withdrawal produces differential effects on the rate of lever-pressing for brain self-stimulation in the hypothalamus and midbrain in rats

    Pharmacol. Biochem. Behav

    (1983)
  • G.J. Schaefer et al.

    Changes in response rates and reinforcement thresholds for intracranial self-stimulation during morphine withdrawal

    Pharmacol. Biochem. Behav

    (1986)
  • P. Schnur et al.

    Blocking naloxone-precipitated withdrawal in rats and hamsters

    Pharmacol. Biochem. Behav.

    (1992)
  • G. Schulteis et al.

    Clonidine blocks acquisition but not expression of conditioned opiate withdrawal in rats

    Neuropsychopharmacology

    (1998)
  • G. Schulteis et al.

    Anxiogenic-like effects of spontaneous and naloxone-precipitated opiate withdrawal in the elevated plus-maze

    Pharmacol. Biochem. Behav.

    (1998)
  • A. Seillier et al.

    The dual FAAH/MAGL inhibitor JZL195 has enhanced effects on endocannabinoid transmission and motor behavior in rats as compared to those of the MAGL inhibitor JZL184

    Pharmacol. Biochem. Behav.

    (2014)
  • L. Stinus et al.

    Nucleus accumbens and amygdala are possible substrates for the aversive stimulus effects of opiate withdrawal

    Neuroscience

    (1990)
  • T. Sugiura et al.

    2-Arachidonoyglycerol: a possible endogenous cannabinoid receptor ligand in brain

    Biochem. Biophys. Res. Comm.

    (1995)
  • T. Suzuki et al.

    Modification of the effects of naloxone in morphine-dependent mice

    Life Sci.

    (1989)
  • P. Valeri et al.

    Effects of dapiprazole, clonidine and yohimbine on the development of dependence and withdrawal behaviour in mice

    Drug Alcohol Depend.

    (1989)
  • E.J. Van Bockstaele et al.

    Low dose naltrexone administration in morphine dependent rats attenuates withdrawal-induced norepinephrine efflux in forebrain

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2008)
  • D. Vigano et al.

    Molecular and cellular basis of cannabinoid and opioid interactions

    Pharmacol. Biochem. Behav.

    (2005)
  • A. Wilson-Poe et al.

    Distribution of CB1 cannabinoid receptors and their relationship with mu-opioid receptors in the rat periaqueductal gray

    Neuroscience

    (2012)
  • Z. Zhang et al.

    Withdrawal from acute morphine dependence is accompanied by increased anxiety-like behavior in the elevated plus maze

    Pharmacol. Biochem. Behav.

    (2008)
  • L. Alguacil et al.

    An automated method for the evaluation of jumping activity in mice. Effects of clonidine on morphine withdrawal

    Methods Find. Exp. Clin. Pharmacol.

    (1989)
  • A.A. Altarifi et al.

    Some determinants of morphine effects on intracranial self-stimulation in rats: dose, pretreatment time, repeated treatment, and rate dependence

    Behav. Pharmacol.

    (2011)
  • I. Broseta et al.

    Isolation decreases physical and motivational aspects of morphine withdrawal

    Behav. Pharmacol.

    (2005)
  • S. Caillé et al.

    Total neurochemical lesion of noradrenergic neurons of the locus ceruleus does not alter either naloxone-precipitated or spontaneous opiate withdrawal nor does it influence ability of clonidine to reverse opiate withdrawal

    J. Pharmacol. Exp. Ther.

    (1999)
  • S.R. Childers et al.

    Opioid and cannabinoid receptor inhibition of adenylyl cyclase in brain

    Ann. N.Y. Acad. Sci.

    (1992)
  • T.J. Cicero et al.

    Diversion and abuse of methadone prescribed for pain management

    JAMA

    (2005)
  • B.F. Cravatt et al.

    Supersensitivity to anandamide and enhanced endogenous cannabinoid signaling in mice lacking fatty acid amide hydrolase

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

    (2001)
  • Cited by (29)

    • Alleviation of opioid withdrawal by cannabis and delta-9-tetrahydrocannabinol: A systematic review of observational and experimental human studies

      2022, Drug and Alcohol Dependence
      Citation Excerpt :

      This strategy is dovetailed by a wealth of evidence showing crosstalk between the opioid and cannabinoid receptor systems, which overlap at the anatomical, neurochemical, and behavioral levels (Scavone et al., 2013a). Several preclinical studies demonstrate that exogenous delta-9-tetrahydrocannabinol (THC), the main psychoactive constituent of cannabis, reduces opioid withdrawal in opioid-dependent animals (Bhargava, 1976a, 1976b; Cichewicz and Welch, 2003; Gamage et al., 2015; Hine et al., 1975a, 1975b; Lichtman et al., 2001); and, that, conversely, cannabinoid receptor antagonists/inverse agonizts (e.g., rimonabant) precipitate opioid withdrawal (Dunn et al., 2019; Scavone et al., 2013a). Convergent lines of evidence also show that cannabinoid agonizts may alleviate pain (Finn et al., 2021; Wang et al., 2021), an important feature of opioid withdrawal — especially when OUD and chronic pain co-occur (Coloma-Carmona et al., 2019; Rodriguez-Espinosa et al., 2021).

    • The impact of naturalistic cannabis use on self-reported opioid withdrawal

      2020, Journal of Substance Abuse Treatment
      Citation Excerpt :

      Substantial preclinical research implicates the endocannabinoid system as having a role in the expression of opioid withdrawal. Indeed, administering exogenous cannabinoids, delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD), and increasing the endogenous cannabinoid anandamide, have all been shown to decrease opioid withdrawal severity in preclinical models (Bhargava, 1976; Cichewicz, 2004; Gamage et al., 2015; Hine, Friedman, Torrelio, & Gershon, 1975; Hine, Torrelio, & Gershon, 1975a, 1975b; Lichtman, Sheikh, Loh, & Martin, 2001; Ramesh et al., 2013; Vela, Ruiz-Gayo, & Fuentes, 1995). Very little human research on this topic exists.

    • Arachidonoyl serotonin (AA-5-HT) modulates general fear-like behavior and inhibits mesolimbic dopamine release

      2019, Behavioural Brain Research
      Citation Excerpt :

      Most drugs of abuse enhance phasic dopamine release in the NAc which is the central mechanism for the reinforcing properties of these drugs [82]. Therefore, our data suggest that AA-5-HT administration may not exert reinforcing effects, which is similar to research indicating a lack of reinforcing properties of FAAH inhibitors in other animal models [83–85]. Together, our results suggest that systemic AA-5-HT administration elicited strain-dependent patterns of attenuated evoked mesolimbic dopamine release.

    View all citing articles on Scopus
    View full text