49
Views
18
CrossRef citations to date
0
Altmetric
Review

Role of endocannabinoids in regulating drug dependence

, , &
Pages 711-721 | Published online: 15 Jan 2008

Abstract

This review will discuss the latest knowledge of how the endocannabinoid system might be involved in treating addiction to the most common illicit drugs. Experimental models are providing increasing evidence for the pharmacological management of endocannabinoid signaling not only to block the direct reinforcing effects of cannabis, opioids, nicotine and ethanol, but also for preventing relapse to the various drugs of abuse, including opioids, cocaine, nicotine, alcohol and metamphetamine. Preclinical and clinical studies suggest that the endocannabinoid system can be manipulated by the CB1 receptor antagonist SR141716A, that might constitute a new generation of compounds for treating addiction across different classes of abused drugs.

Introduction

Drug addiction is a chronic relapsing brain disorder, manifested as an intense desire for the drug, with impairment of the ability to control the urges to take the drug, even at the expense of serious adverse consequences (CitationCamì and Farré 2003). These behavioral abnormalities develop gradually with repeated exposure to a drug of abuse, and can persist for months or years after discontinuation of use, suggesting that addiction can be considered a form of drug-induced neural plasticity (CitationNestler 2004).

Several compounds can lead to addictive behavior including opioids, psychostimulants, cannabinoids, alcohol and nicotine, and although their initial mechanism of action affects different neurochemical targets, the resulting neural dysregulation involves similar neurochemical and neuroanatomical pathways (CitationHyman and Malenka 2001). The limbic component of basal ganglia pathways, the endogenous opioid system and the brain-pituitary stress system are all essential for the addictive properties of most drugs of abuse, whose interaction with these circuits leads to a common dysregulation of brain motivational and reward pathways (CitationMaldonado et al 2006).

The limbic component of the basal ganglia pathway is a common neuronal substrate for the reinforcing properties of drugs of abuse and drives the motivational, emotional and affective information on behavior (see for review CitationKoob 1992; CitationDi Chiara 1999; CitationKoob et al 2004; CitationPierce et al 2006). The mesocorticolimbic dopaminergic pathway (comprising dopaminergic neurons in the ventral tegmental area – VTA – innervating the nucleus accumbens – NAc – hippocampus, amygdala, medial prefrontal cortex and ventral pallidum), is a vital factor governing the fl ow of information through the limbic circuit comprising the interconnected nuclei. Thus dopamine is considered one of the most important actors in the rewarding effects of drugs of abuse, as suggested by the finding that most of the drugs abused by humans raise dopamine levels in the NAc, and blockade of dopamine transmission reduces the rewarding effect of psychostimulants (see for review CitationPierce and Kumaresan 2006). Moreover, mesolimbic dopaminergic neurons communicate with cerebral areas involved in cognitive functions and dopamine release in the forebrain can be considered a learning signal. In the NAc glutamatergic projections from the cerebral cortex, amygdala and hippocampus drive information about external situations and internal emotional and physiological states, thus contributing to addiction by consolidating reward-driven behavior (CitationHyman and Malenka 2001; CitationKauer 2004).

The endocannabinoid system and addictive behavior

Besides the importance of the mesocorticolimbic dopaminergic system in addiction, the shared mechanisms in the development of addictive behavior have not yet been fully identified so this review will focus on recent findings pointing towards a role of the endocannabinoid system in the circuitry underlying drug addiction.

Knowledge of the endocannabinoid system has been largely boosted since the CB1 receptor was cloned in 1990 and we now understand that the endocannabinoid system consists of cannabinoid receptors, endogenous ligands and several proteins responsible for their synthesis and degradation (see for review CitationBisogno et al 2005). Two cannabinoid receptors, CB1 and CB2 have been cloned and characterized, both belonging to the class of G protein-coupled receptors. CB1 has been located in the central nervous system and peripheral tissues and CB2 appeared mainly in the cells of the immune system (CitationFride and Mechoulam 2003) although it has now also been identified in brainstem, cortex and cerebellum neurons (CitationVan Sickle et al 2005).

The most fully characterized endocannabinoid substances isolated from brain tissue are anandamide (AEA) and 2-arachidonylglycerol (2-AG) (CitationFride and Mechoulam 2003). Endocannabinoids serve as neuromodulators in many physiological processes and once released from postsynaptic neurons upon depolarization, they activate presynaptic receptors, resulting in inhibition of the release of both excitatory and inhibitory transmitters (see for review CitationFride 2005). In this capacity the endocannabinoid system may have important additional roles in the regulation of synaptic brain function.

CB1 receptors are abundant in the brain reward circuitry, and the dopaminergic neurons of the mesocorticolimbic pathway are regulated by excitatory and inhibitory inputs influenced by activation of cannabinoid receptors (see for review CitationGardner 2005). Endocannabinoids released after depolarization in the NAc and from dopaminergic neurons in the VTA may possibly influence GABAergic and glutamatergic afferents by acting as retrograde messengers on CB1 receptors.

CitationWenger et al (2003) reported the presence of cannabinoid receptors in tyrosine hydroxylase-expressing neurons (most probably dopaminergic neurons) of the NAc, VTA, striatum and pyriform cortex, suggesting the endocannabinoid system might directly influence dopaminergic reward mechanisms. CB1 receptors are present in other areas related to reward and motivation (such as the basolateral amygdala and hippocampus) and endocannabinoids induce long-term depression (LTD) of the inhibitory synapses in the hippocampus, contributing to the synaptic plasticity involved in the learning processes related to addictive behavior (CitationDe Vries and Schoffelmeer 2005).

The endocannabinoid system is certainly the primary site of action for cannabinoid addiction and in fact cannabinoids, like other drugs of abuse, induce tolerance and physical dependence and activate a rewarding system (see for review CitationParolaro et al 2005; CitationFattore et al 2005; CitationGonzalez et al 2005). The exact sites and substrates of cannabinoid action in the core VTA-medial forebrain bundle (MFB)-NAc reward axis and on reward-related behaviors are still not clear but there is evidence that cannabinoids enhance brain reward substrates, acting on both dopamine-dependent substrates in the VTA and dopamine-dependent/independent ones in the NAc (CitationLupica et al 2004).

However, the endocannabinoid system certainly has an overall effect on the reward circuitry and participates in the rewarding and addictive properties of all prototypical drugs of abuse such as opioids, nicotine, alcohol and psychostimulants (cocaine and amphetamine). Animal models of drug reward provide evidence of the endogenous cannabinoids’ role in the rewarding effects of several addictive drugs, and pharmacological manipulation of endocannabinoid tone with SR141716A (rimonabant, a specific CB1 receptor antagonist) in humans gave positive results (CitationAnthenelli and Despres 2004).

Two complementary approaches have been used to demonstrate the endocannabinoid system’s role in addictive behavior; the first is a genetic approach evaluating changes to the addictive properties of several drugs of abuse in CB1 knockout mice, and the second is a pharmacological approach looking at the effect of SR141716A on drugs’ addictive properties. Research in this field has also gained from the use of validated experimental models for the subjective effects of drugs (drug discrimination), their rewarding/reinforcing properties (intravenous self-administration, conditioned place preference – CPP – and intracranial self-stimulation), the influence of environmental factors on drug-seeking behavior (CPP, second-order schedules of self-administration, reinstatement of extinguished drug-seeking behavior and other relapse models), and the withdrawal states associated with abrupt termination of drug action (administration of a selective antagonist after chronic exposure).

The endocannabinoid system in opioid addiction

Opioids and cannabinoids have several similar pharmacological effects, including analgesia and stimulation of brain circuitry, that are believed to underlie drug addiction and reward. There is ample evidence of a role for the endocannabinoid system in opioid dependence (). Cannabinoids attenuated morphine and methadone withdrawal signs (CitationHine et al 1975; CitationDeikel and Carder 1976; CitationVela et al 1995; CitationYamaguchi et al 2001; CitationDel Arco et al 2002) and the cannabinoid antagonist SR141716A precipitated abstinence in morphine-dependent rats (CitationNavarro et al 1998; CitationMaldonado 2002). The severity of naloxone-precipitated morphine withdrawal was robustly attenuated in CB1 ko mice (CitationLedent et al 1999; CitationLichtman et al 2001) and long-term treatment with SR141716A reduced the intensity of naloxone-precipitated opioid withdrawal (CitationRubino et al 2000; CitationMas Nieto et al 2001); however an acute dose of SR141716A just before naloxone did not affect the incidence of withdrawal signs (CitationMas-Nieto et al 2001).

Table 1 Cannabinoid system in opioid addiction

Thus it appears that chronic blockade of CB1 receptors is needed to alleviate the main signs of morphine abstinence, suggesting that chronic treatment with SR141716A might be useful in the opiate withdrawal syndrome. In addition changes in CB1 receptor function (in terms of receptor binding and coupling with G protein) and/or in endocannabinoid levels were observed in specific brain areas of morphine-dependent animals, although the results ranged from no change to a decrease or even an increase (CitationRomero et al 1998; CitationGonzalez et al 2003; CitationViganò et al 2005).

The opioid antagonist naloxone precipitated abstinence symptoms in rats tolerant to Δ9-tetrahydrocannabinol (Δ9-THC) (CitationKaymakcalan et al 1977) and SR141716A-precipitated withdrawal was dose-dependently reduced by morphine (CitationLichtman et al 2001). The somatic expression of cannabinoid withdrawal was attenuated in mice lacking pre-pro-enkephalin or mu opioid receptor genes (CitationValverde et al 2000; CitationLichtman et al 2001) whereas the deletion of mu, kappa and delta opioid receptors did not affect cannabinoid withdrawal (CitationGhozland et al 2002). In contrast, in double ko mice deficient in mu and delta opioid receptors, cannabinoid withdrawal was significantly reduced (CitationCastane et al 2003), suggesting that a cooperative action of both receptors was required for the expression of Δ9-THC dependence.

The traditional animal paradigm of drug dependence further confirmed the importance of the endocannabinoid system in opioid addiction. CitationManzanedo et al (2004) reported that pre-exposure to the synthetic cannabinoid agonist WIN55212-2 increased the rewarding effect of morphine evaluated in a place preference paradigm, supporting the idea that using cannabis might make an individual more vulnerable to opiate addiction. More recently, Goldberg’s group (CitationSolinas et al 2005) reported that the reinforcing efficacy of heroin, measured in a progressive ratio schedule of i.v. heroin self-administration, was significantly enhanced by Δ9-THC and WIN55212-2 but not by compounds that raise the levels of endocannabinoids by blocking their uptake or metabolism; this suggested that the interaction between cannabinoids and opioids might be mediated by their receptors and signaling pathways rather than by opioid-induced release of endogenous cannabinoids.

In contrast, approaches involving the CB1 receptor antagonist SR141716A suggested the endocannabinoids had a facilitating effect on opioid reinforcement that was unmasked by CB1 receptor blockade. SR141716A reduced opioid self-administration and conditioned place preference in rodents (CitationChaperon et al 1999; CitationBraida et al 2001; CitationNavarro et al 2001, Citation2004; CitationMas Nieto et al 2001; CitationCaille and Parsons 2003; CitationDe Vries et al 2003; CitationFattore et al 2003; see for review CitationFattore et al 2005 and CitationMaldonado et al 2006). SR141716A had more effect on heroin self-administration when more effort was required to obtain a heroin infusion; for example it was efficacious under a progressive ratio schedule of reinforcement (high price of drug), weaker under a fixed ratio schedule 5 (low price of drug) and null under a fixed ratio schedule 1 (very low price) (CitationSolinas et al 2003).

These results strengthen the idea that SR141716A attenuates the reinforcing effects of heroin and provide support for the potential efficacy of cannabinoid CB1 antagonists in the prevention and treatment of opioid reward. Thus, morphine conditioned place preference (CitationMartin et al 2000) and self-administration (CitationLedent et al 1999; CitationCossu et al 2001) were abolished in CB ko mice although the results on morphine CPP in CB1 ko mice tended to vary. CitationMartin et al (2000) found morphine induced CPP in wild-type mice but there was no such response in ko mice, indicating that the drug had no rewarding effects in the absence of CB1 cannabinoid receptors. CitationRice et al in 2002, reported that CB1 receptor ko mice developed a strong place preference to morphine, similar to that in wild-type Swiss-Webster mice; this indicated that the brain cannabinoid system made no contribution to morphine reward.

One explanation of these conflicting results might be found in the slightly more intensive conditioning paradigm and differences in the conditioning chambers used in the last study.

In summary, the cannabinoid system plays a permissive role in opioid motivational and rewarding properties. The rewarding effects of Δ9-THC were suppressed in opioid-receptor ko mice (CitationGhozland et al 2002; CitationCastane et al 2003) and attenuated by opioid antagonists (CitationBraida et al 2001; CitationJustinova et al 2004).

It is interesting that SR141716A did not modify the dopamine releasing effect of heroin in the NAc (CitationTanda and Di Chiara 1997; CitationCaille and Parsons 2003). CitationCaille and Parsons (2006) have subsequently focused on an additional substrate in opiate reward, namely the ventromedial pallidum (VP), a cerebral area that receives dense GAB-Aergic input from the NAc. Opiates strongly reduced VP GABA efflux and the resulting disinhibition of the VP is thought to contribute to the positive reinforcing effects of opiates (CitationCaille and Parsons 2004). SR141716A caused a dose-dependent blockade of the effect of morphine on VP GABA efflux without influencing morphine’s dopamine-releasing effect. However, SR14716A did not alter cocaine self-administration, or cocaine-induced decrements in VP GABA efflux and increases in NAc dopamine. This is consistent with evidence that selective inactivation of CB1 receptors reduces opiate-, but not psychostimulant-maintained self-administration. However, the CB1 receptor agonist WIN55212-2 reduced VP GABA efflux in a manner similar to morphine, and this effect was reversed by the opiate receptor antagonist naloxone. These results point to an interaction between cannabinoids and opioids in their effects on VP activity, and suggest that SR141716A attenuates opiate reward by reducing the opiates’ inhibitory influence on NAc medium spiny neurons.

Finally, endocannabinoid tone seems to have a particularly interesting role in relapse to opiate abuse, especially in humans. Detoxification from opiate addiction has been a medical problem for as long as opiates have been abused, as relapses occur even after long drug-free periods. The endocannabinoid system almost certainly plays a part in triggering or preventing reinstatement of drug-seeking behavior (see for review CitationFattore et al 2006).

The three synthetic CB1 receptor agonists WIN55212-2, CP55940 and HU210 promptly reinstate heroin-seeking after a long drug-free period (CitationDe Vries et al 2003; CitationFattore et al 2003). Rimonabant, however, attenuates heroin-induced reinstatement of heroin-seeking behavior (CitationDe Vries et al 2003; CitationFattore et al 2003) suggesting that CB1 receptor blockade alters heroin’s reinforcing consequences.

Further supporting the notion of a close reciprocal relationship between cannabinoid and opioid systems in relapse, blockade of opioid receptors by naloxone prevented relapse to cannabinoids (CitationSpano et al 2004) and a priming injection of heroin reinstated cannabinoid-seeking behavior after three weeks of extinction. Interestingly, SR141716A per se did not reinstate responding but did prevent cannabinoid-seeking behavior triggered by heroin (CitationSpano et al 2004).

The endocannabinoid system in nicotine addiction

There appears to be a functional interaction between the endogenous cannabinoid system and nicotine addiction (). In the mouse CPP paradigm, co-administration of sub-threshold doses of Δ9-THC and nicotine induced rewarding effects (CitationValjent et al 2002). Acute Δ9-THC significantly lowered the incidence of several precipitated nicotine withdrawal signs and improved the aversive motivational consequences of nicotine withdrawal in mice (CitationBalerio et al 2004). These findings suggest that each drug enhances the rewarding effect of the other, and that cannabis might be used to reduce aversive reactions resulting from nicotine withdrawal. In contrast, CitationValjent et al (2002) showed an enhancement in the somatic expression of withdrawal in animals co-treated with nicotine and Δ9-THC, suggesting an asymmetric relationship between the two compounds. Nicotine had no rewarding effect in CB1 ko mice in a place preference paradigm (CitationCastane et al 2002). By contrast, CitationCossu et al (2001) reported that the absence of CB1 receptors did not affect nicotine self-administration. Methodological aspects such as the specific paradigm and the dose used may partly explain this discrepancy.

Table 2 Cannabinoid system in nicotine and alcohol addiction

The second approach used to demonstrate the endocannabinoid system’s role in nicotine addiction involves SR141716A. Pretreatment with this specific CB1 receptor antagonist reduced nicotine self-administration, nicotine-seeking behavior induced by conditioned cues in rats (CitationLe Foll and Goldberg 2004; CitationCohen et al 2005), nicotine-induced dopamine release in the NAc (CitationCohen et al 2002) and the dopaminergic component of nicotine discrimination (CitationCohen et al 2002).

To conclude, since dopamine release in the NAc is thought to play a major role in the positive reinforcement of nicotine, and although findings in CB1 receptor ko mice are inconsistent, SR141716A may well have some antismoking activity, by reducing the rewarding/reinforcing effect of nicotine.

Some recent papers have looked at the utility of SR141716A for reducing cues associated with nicotine-seeking behavior, one of the major causes of relapse in former smokers. CitationForget et al (2005) showed that SR141716A impaired both the establishment and the short-term expression of CPP induced by nicotine, suggesting that endogenous cannabinoids are implicated in nicotine’s motivational effects. Interestingly, rimonabant did not affect the long-term expression of the incentive learning, suggesting other cannabinoid-independent mechanisms are involved (CitationForget et al 2005). However, CitationCohen et al (2005) found that rimonabant attenuated the long-term influence of environmental stimuliresponsible for relapse in nicotine-seeking behavior. After demonstrating the persistence of conditioned behavior in response to nicotine-related cues several weeks after nicotine withdrawal, Cohen et al showed that rimonabant could reduce the responding maintained by these cues three months after stopping smoking. Thus rimonabant may not only help stop people smoking but may also help them remain abstinent.

Biochemical investigations found altered levels of AEA and 2-AG in several brain areas in animals chronically treated with nicotine. AEA was elevated in the limbic area and brainstem, and 2-AG in the brainstem, but one or both were reduced in other regions such as hippocampus, striatum and cerebral cortex. CB1 receptor levels were not altered in any of these brain areas (CitationGonzalez et al 2002; CitationBalerio et al 2004).

To summarize, the pharmacological and cellular studies described indicate that the endogenous cannabinoid system has a specific role in nicotine responses related to its addictive behavior and open up new possibilities for the treatment of this major public health disorder.

There are already some preliminary data from the STRATUS-US trial (smoking cessation in smokers motivated to quit) on the efficacy of rimonabant in humans (CitationAnthenelli and Despres 2004). This clinical trial enrolled 787 smokers in the United States. The participants were randomized to rimonabant 5 mg or 20 mg, or placebo. The study lasted ten weeks and the smokers were allowed to smoke during the first two weeks but were asked to abstain after this. The quitting rate in the 2-mg rimonabant group was double that with placebo and they showed markedly less weight gain during the ten-week study period (CitationAnthenelli and Despres 2004).

The endocannabinoid system in alcohol addiction

Recent studies suggest that the endocannabinoid system has a major part among the neurotransmitter systems involved in regulating different alcohol-related phenomena, including tolerance, vulnerability, reinforcement, consumption and metabolism (). Thus acute administration of synthetic or endogenous cannabinoid agonists stimulated alcohol intake in Sardinian alcohol-preferring rats (CitationColombo et al 2002) and C57BL/6J mice (CitationWang et al 2003), and dose-dependently increased break points (an indicator of motivation to drink alcohol) for beer in Wistar rats (CitationGallate et al 1999). All these effects were completely prevented by pre-treatment with the cannabinoid antagonist SR141716A. Furthermore, genetic deletion of the FAAH enzyme (CitationBasavarajappa et al 2006) or an injection of URB597 (an irreversible FAAH inhibitor) into the prefrontal cortex enhanced motivation to drink alcohol (CitationHansson et al 2006), pointing clearly to a role for the endocannabinoid system in alcohol addiction. It is in fact now established that SR141716A, given alone, has effects on alcohol-related behavior opposite to those of the CB1 receptor agonists. For example, SR141716A prevented the acquisition of alcohol drinking behavior in alcohol-naïve Sardinian alcohol-preferring rats with a free choice between alcohol (10%, v/v) and water (CitationSerra et al 2001). It also reduced ethanol consumption in C57Bl/6 mice at doses only marginally affecting regular chow intake or water drinking; this suggests that the endogenous cannabinoid system may affect the appetitive value of ethanol (CitationArnone et al 1997).

Similar reductions in voluntary alcohol intake were obtained in Sardinian alcohol-preferring rats (CitationColombo et al 1998), and Wistar rats (CitationLallemand et al 2001) after chronic alcoholization (a model of the “maintenance” phase of human alcoholism). In the Sardinian alcohol-preferring rats the CB1 receptor antagonist completely abolished the effect of alcohol deprivation (ie, the temporary increase in alcohol intake after a period of withdrawal, a model for relapse episodes in human alcoholics). This suggested that the cannabinoid CB1 receptor might be part of the neural substrate of the alcohol deprivation effect and that SR141716A may have anti-relapse properties (CitationSerra et al 2002).

Finally, SR141716A reduced oral self-administration and attenuated the appetitive properties of alcohol in unselected rats under operant procedures (CitationGallate and McGregor 1999; CitationFreedland et al 2001; CitationColombo et al 2004). These data were confirmed by CitationGessa et al (2005) using a second CB1 receptor antagonist, the newly synthesized SR147778, which suppressed acquisition and maintenance of alcohol drinking, relapse-like drinking and motivation to consume alcohol in Sardinian alcohol-preferring rats.CitationCippitelli et al (2005) provided clear evidence that blockade of CB1 receptors reduced both ethanol self-administration and conditioned reinstatement of alcohol-seeking behavior in Marchigian-Sardinian alcohol-preferring rats and Wistar rats, the genetically selected rats showing higher sensitivity to rimonabant. These researchers also reported that at least in the strain of rats bred for its ethanol preference, the Marchigian-Sardinian alcohol-preferring rats, CB1 cannabinoid receptor mRNA expression was increased in brain areas relevant for processing reward and reward-associated behavior, suggesting that alterations in the function of the CB1 receptor system may be linked to genetic vulnerability to alcohol misuse (CitationCippitelli et al 2005).

All these results further reinforce the concept that pharmacological blockade of the CB1 receptor may offer a novel approach to the treatment of alcoholism, not only for consumption but also for context-induced relapses to alcohol, one of the main problems in the treatment of this disorder.

The CB1 receptor signaling system’s participation in alcohol drinking and alcohol sensitivity was confirmed using CB1 receptor ko mice. CB1 −/− mice with CD1 background showed lower ethanol intake and less preference, effects associated with dramatic sensitivity to the hypothermic and hypolocomotor effects in response to low doses of ethanol (CitationNaassila et al 2004). These mice also had more severe withdrawal-induced convulsions. Since previous studies in rodents have suggested that high levels of ethanol drinking are often associated with resistance to its intoxicant effects (CitationSchuckit 1994; CitationKurtz et al 1996), the lower ethanol consumption in CB1 ko mice might be due to their greater sensitivity to ethanol’s acute effects.

Chronic in vivo (CitationGonzalez et al 2002) or in vitro (CitationBasavarajappa and Hungund, 1999a; CitationBasavarajappa et al 2003) ethanol exposure increased accumulation of AEA. This was inhibited by pertussis toxin and the CB1 receptor antagonist SR141716A, and paralleled by the activation of Ca2+-dependent and arachidonic acid-specific phospholipase A2, a key enzyme in the formation of endocannabinoids in neuronal cells and brain (CitationBasavarajappa et al 1997, Citation1998). The mechanism by which chronic ethanol exposure increases the endocannabinoid content remains to be established, though CitationBasavarajappa et al (2003) have reported that in cerebellar granular neurons chronic exposure to alcohol led to an increase in extracellular AEA by inhibiting its uptake. This event was CB1 receptor-independent since it also occurred in CB1 ko mice and cannabinoid CB1 receptor antagonists did not alter the effects of chronic ethanol on AEA transport. Moreover, in rodents chronic exposure to alcohol impairs CB1 receptor function, lowering the levels of CB1 receptor binding, expression and CB1 receptor/G protein coupling (CitationBasavarajappa and Hungund 1999b; CitationOrtiz et al 2004). This might be due to overstimulation of receptors through increased endocannabinoid synthesis. These studies suggest that during the development of alcohol tolerance there are changes in the steady state in the endogenous cannabinoid system, and this might explain the altered response to alcohol.

These converging findings suggest that cannabinoid CB1 receptor blockade may be an effective therapeutic approach for alcohol dependence in humans but information on the clinical efficacy of rimonabant is still lacking.

The endocannabinoid system in addiction to psychostimulants

Psychostimulants differ from the drugs of abuse since they affect mesolimbic dopaminergic terminals directly, raising dopamine levels in the NAc by direct action on dopaminergic axon terminals; the other drugs seem to induce rewarding effects by increasing dopaminergic neuron firing rates, acting in the VTA, possibly through the release of endocannabinoids (CitationLupica and Riegel 2005).

This may help us understand why endocannabinoids are not involved in cocaine’s or amphetamine’s primary reinforcing effects. In fact, there are several reports () that genetic deletion or pharmacological blockade of CB1 receptors does not alter cocaine or amphetamine self-administration (CitationCossu et al 2001; CitationDe Vries et al 2001; CitationBraida et al 2005; CitationLesscher et al 2005) or the neurochemical correlate of this behavior, namely dopamine release in the NAc (Cossu, unpublished results, CitationSoria et al 2005). Similarly, cocaine-induced CPP was not modified in CB1 ko mice (CitationMartin et al 2000; CitationHouchi et al 2005). These results clearly indicate that CB1 receptors are probably not involved in the primary reinforcing effects of psychostimulants.

Table 3 Cannabinoid system in psychostimulant addiction

Although acute reinforcing properties are essential for the establishment of drug addiction, other complex behavioral processes are involved in consolidating this chronic relapsing disorder (CitationKoob and LeMoal 2001), so the acute reinforcing effects of the drug are only the first step in the acquisition of stable operant self-administration responding. Besides the mesolimbic dopaminergic system, particularly the NAc, it has been proposed that dopamine-independent neuronal circuits are involved in regulating reward-related processes (CitationLupica et al 2004). CB1 cannabinoid receptors are highly expressed in other brain regions involved in the rewarding circuitry such as the basolateral amygdala, medial prefrontal cortex, and hippocampus, so mechanisms involving CB1 receptors in these structures might well participate in other aspects such as consolidation and relapse of cocaine-seeking behavior. In line with this hypothesis, CitationSoria et al (2005) found that only 25% of CB1 ko mice, compared with 75% of the wild-type, acquired reliable operant responding to self-administer the most effective dose of cocaine (1 mg/kg/infusion), and needed more sessions to attain this behavior. Moreover, the maximal effort to obtain a cocaine infusion was significantly lower after genetic ablation of CB1 receptors, indicating decreased motivation for maintaining cocaine-seeking behavior. Results were similar after pharmacological blockade of CB1 receptors with SR141716A in wild-type litter mates (CitationSoria et al 2005). The lack of motivation for cocaine in CB1 ko or SR141716A-pretreated mice might be due to impaired detection, association, and representation of the reward signal or to inadequate responding to the rewarding stimuli (CitationSoria et al 2005).

In line with the idea of CB1 receptors involved in these other aspects of reward, CitationFattore et al (1999) found that WIN 55212-2 reduced intravenous cocaine self-administration, suggesting that stimulation of CB1 receptors may potentiate cocaine’s reinforcing effects. Similar findings were reported with amphetamine (CitationBraida and Sala 2002): the combination of CP-55,940 with the maximal reinforcing unit dose of 3,4-methylendioxymethamphetamine (MDMA) significantly lowered the mean number of drug-associated lever pressings in comparison with the drug alone, suggesting a synergistic action of cannabinoid agonists on MDMA’s reinforcing properties.

Finally, CB1 receptors play an important role in relapse to psychostimulants. CitationDe Vries et al (2001) found that the cannabinoid agonist HU210 provoked relapse to drug-seeking in animals after prolonged withdrawal from cocaine self-administration, whereas blockade of the CB1 receptor by SR141716A attenuated the relapse induced by re-exposure to cocaine-associated cues or the drug itself. CitationXi et al (2006) have now shown that the CB1 antagonist AM251, administered systemically, selectively inhibited cocaine-induced reinstatement of reward-seeking behavior by a glutamate-dependent mechanism. CB1 receptor-mediated disinhibition of NAc glutamate release could activate presynaptic mGluR2/3 receptors, which then inhibited cocaine-enhanced NAc glutamate release and cocaine-triggered reinstatement of drug-seeking behavior (CitationXi et al 2006). Along the same lines, SR141716A blocked the reinstatement of methamphetamine-seeking behavior in rats (CitationAnggadidiredja et al 2004).

Given the paucity of effective medications to treat psychostimulant addiction, and although the endocannabinoid system does not participate in the primary reinforcing effects of this class of drugs, CB1 receptor antagonists may offer hope for treating consolidated psychostimulant-seeking behavior and relapse.

Conclusions and future directions

In the last 25 years the neurobiological and behavioral mechanisms that lead to drug dependence have been extensively investigated but clinical treatment is still unsatisfactory and ineffective in many subjects.

Experimental models are now providing evidence for the pharmacological management of endocannabinoid signaling not only to block the direct reinforcing effects of cannabis, opioids, nicotine and ethanol, but also to prevent relapse to these various substances of abuse, also including cocaine and metamphetamine. The endocannabinoid system can be manipulated by SR141716A and by all the new compounds that protect AEA and 2-AG from deactivation and prolong the lifespan of these endocannabinoid substances in vivo. Rimonabant reduces the motivational effect of drug-related stimuli and drug re-exposure, probably by altering synaptic plasticity, thus providing an effective means of preventing relapse and a new tool for the treatment of drug abuse.

Although further studies are needed to clarify the precise mechanism underlying the endocannabinoid system’s role in addiction, some promising clinical findings have now been presented (eg, smoking cessation). A new question has now arisen from the discovery of CB2 receptors in the brain: are they involved in drug addiction?

References

  • AnggadiredjaKNakamichiMHiranitaT2004Endocannabinoid system modulates relapse to methamphetamine seeking: possible mediation by the arachidonic acid cascadeNeuropsychopharmacology291470815085091
  • AnthenelliRMDespresJP2004Effects of Rimonabant in the reduction of major cardiovascular risk factors Results from the STRATUS-US trial (smoking cessation in smokers motivated to quit)In american college of cardiology 53rd annual scientific session2004, Mar 7–10New Orleans, LA
  • ArnoneMMaruaniJChaperonF1997Selective inhibition of sucrose and ethanol intake by SR141716, an antagonist of central cannabinoid (CB1) receptorsPsychopharmacology13210469272766
  • BalerioGNAsoEBerrenderoF2004Delta9-tetrahydrocannabinol decreases somatic and motivational manifestations of nicotine withdrawal in miceEur J Neurosci2027374815548217
  • BasavarajappaBSSaitoMCooperTB1997Activation of arachidonic acid-specific phospholipase A2 in human neuroblastoma cells after chronic alcohol exposure: prevention by GM1 gangliosideAlcohol Clin Exp Res2111992039347079
  • BasavarajappaBSCooperTBHungundBL1998Effect of chronic ethanol exposure on mouse brain arachidonic acid specific phospholipase A2Biochem Pharmacol55515219514087
  • BasavarajappaBSHungundBL1999aChronic ethanol increases the cannabinoid receptor agonist anandamide and its precursor N-arachidonoyl-phosphatidyl-ethanolamine in SK-N-SH cellsJ Neurochem7252289930723
  • BasavarajappaBSHungundBL1999bDown-regulation of cannabinoid receptor agonist-stimulated [35S]GTP gamma S binding in synaptic plasma membrane from chronic ethanol exposed mouseBrain Res81589979974126
  • BasavarajappaBSSaitoMCooperTB2003Chronic ethanol inhibits the anandamide transport and increases extracellular anandamide levels in cerebellar granule neuronsEur J Pharmacol466738312679143
  • BasavarajappaBSYalamanchiliRCravattBF2006Increased ethanol consumption and preference and decreased ethanol sensitivity in female FAAH knockout miceNeuropharmacology508344416448676
  • BisognoTLigrestiADi MarzoV2005The endocannabinoid signalling system: biochemical aspectsPharmacol Biochem Behav812243815935454
  • BraidaDIosueSPegoriniS20053,4-Methylenedioxymethamphetamine-induced conditioned place preference (CPP) is mediated by endocannabinoid systemPharmacol Res511778215629265
  • BraidaDPozziMCavalliniR2001Conditioned place preference induced by the cannabinoid agonist CP 55,940: interaction with the opioid systemNeuroscience104923611457579
  • BraidaDSalaM2002Role of the endocannabinoid system in MDMA intracerebral self-administration in ratsBr J Pharmacol13610899212163340
  • CailleSParsonsLH2003SR141716A reduces the reinforcing properties of heroin but not heroin-induced increases in nucleus accumbens dopamine in ratsEur J Neurosci183145914656311
  • CailleSParsonsLH2004Intravenous heroin self-administration decreases GABA efflux in the ventral pallidum: an in vivo microdialysis study in ratsEur J Neurosci20593615233770
  • CailleSParsonsLH2006Cannabinoid modulation of opiate reinforcement through the ventral striatopallidal pathwayNeuropsychopharmacology318041316123766
  • CamiJFarreM2003Drug addictionN Engl J Med3499758612954747
  • CastaneAValjentELedentC2002Lack of CB1 cannabinoid receptors modifies nicotine behavioural responses, but not nicotine abstinenceNeuropharmacology438576712384171
  • CastaneARobledoPMatifasA2003Cannabinoid withdrawal syndrome is reduced in double mu and delta opioid receptor knockout miceEur J Neurosci17155912534979
  • ChaperonFThiebotMH1999Behavioral effects of cannabinoid agents in animalsCrit Rev Neurobiol132438110803637
  • CippitelliABilbaoAHanssonAC2005Cannabinoid CB1 receptor antagonism reduces conditioned reinstatement of ethanol-seeking behavior in ratsEur J Neurosci2122435115869521
  • CohenCPerraultGGriebelG2005Nicotine-associated cues maintain nicotine-seeking behavior in rats several weeks after nicotine withdrawal: reversal by the cannabinoid (CB1) receptor antagonist, Rimonabant (SR141716)Neuropsychopharmacology301455515292905
  • CohenCPerraultGVoltzC2002SR141716, a central cannabinoid (CB(1)) receptor antagonist, blocks the motivational and dopamine-releasing effects of nicotine in ratsBehav Pharmacol134516312394421
  • ColomboGAgabioRFaM1998Reduction of voluntary ethanol intake in ethanol-preferring sP rats by the cannabinoid antagonist SR141716Alcohol Alcohol33126309566474
  • ColomboGSerraSBrunettiG2002Stimulation of voluntary ethanol intake by cannabinoid receptor agonists in ethanol-preferring sP ratsPsychopharmacology159181711862347
  • ColomboGVaccaGSerraS2004Suppressing effect of the cannabinoid CB1 receptor antagonist, SR141716, on alcohol’s motivational properties in alcohol-preferring ratsEur J Pharmacol4981192315363985
  • CossuGLedentCFattoreL2001Cannabinoid CB1 receptor knockout mice fail to self-administer morphine but not other drugs of abuseBehav Brain Res11861511163634
  • De VriesTJHombergJRBinnekadeR2003Cannabinoid modulation of the reinforcing and motivational properties of heroin and heroin-associated cues in ratsPsychopharmacology168164912669182
  • De VriesTJShahamYHombergJR2001A cannabinoid mechanism in relapse to cocaine seekingNat Med71151411590440
  • DeikelSMCarderB1976Attentuation of precipitated abstinence in methadone-dependent rats by delta9-THCPsychopharmacol Commun2615959602
  • Del ArcoINavarroMBilbaoA2002Attenuation of spontaneous opiate withdrawal in mice by the anandamide transport inhibitor AM404Eur J Pharmacol454103412409011
  • De VriesTJSchoffelmeerAN2005Cannabinoid CB1 receptors control conditioned drug seekingTrends Pharmacol Sci268420615992935
  • Di ChiaraG1999Drug addiction as dopamine-dependent associative learning disorderEur J Pharmacol375133010443561
  • FattoreLDeianaSSpanoSM2005Endocannabinoid system and opioid addiction: behavioural aspectsPharmacol Biochem Behav813435915935459
  • FattoreLMartellottaMCCossuG1999CB1 cannabinoid receptor agonist WIN 55,212-2 decreases intravenous cocaine self-administration in ratsBehav Brain Res104141611125733
  • FattoreLSpanoMSCossuG2003Cannabinoid mechanism in reinstatement of heroin-seeking after a long period of abstinence in ratsEur J Neurosci171723612752390
  • FattoreLSpanoMSDeianaS2006An endocannabinoid mechanism in relapse to drug seeking: A review of animal studies and clinical perspectivesBrain Res Brain Res Rev[Epub 11 Jul ahead of print doi:10.1016/j.brainresrev.2006.05.003]
  • ForgetBHamonMThiebotMH2005Cannabinoid CB1 receptors are involved in motivational effects of nicotine in ratsPsychopharmacology1817223415986197
  • FreedlandCSSharpeALSamsonHH2001Effects of SR141716A on ethanol and sucrose self-administrationAlcohol Clin Exp Res252778211236843
  • FrideEMechoulamRMaldonadoR2003New advances in the identification and physiological role of the different components of the endogenous cannabinoidi systemMolecular biology of drug addictionHumana Press17398
  • FrideE2005Endocannabinoids in the central nervous system: from neuronal networks to behaviorCurr Drug Targets CNS Neurol Disord46334216375681
  • GallateJEMcGregorIS1999The motivation for beer in rats: effects of ritanserin, naloxone and SR141716Psychopharmacology142302810208323
  • GallateJESaharovTMalletPE1999Increased motivation for beer in rats following administration of a cannabinoid CB1 receptor agonistEur J Pharmacol3702334010334497
  • GardnerE2005Endocannabinoid signaling system and brain reward: emphasis on dopaminePharmacol Biochem Behav812638415936806
  • GessaGLSerraSVaccaG2005Suppressing effect of the cannabinoid CB1 receptor antagonist, SR147778, on alcohol intake and motivational properties of alcohol in alcohol-preferring sP ratsAlcohol Alcohol40465315582988
  • GhozlandSMatthesHWSimoninF2002Motivational effects of cannabinoids are mediated by mu-opioid and kappa-opioid receptorsJ Neurosci2211465411826143
  • GonzalezSCebeiraMFernandez-RuizJ2005Cannabinoid tolerance and dependence: a review of studies in laboratory animalsPharmacol Biochem Behav813001815919107
  • GonzalezSFernandez-RuizJSparpaglioneV2002Chronic exposure to morphine, cocaine or ethanol in rats produced different effects in brain cannabinoid CB(1) receptor binding and mRNA levelsDrug Alcohol Depend66778411850139
  • GonzalezSSchmidPCFernandez-RuizJ2003Region-dependent changes in endocannabinoid transmission in the brain of morphine-dependent ratsAddict Biol81596612850774
  • HanssonACBermudez-SilvaFJMalinenH2006Genetic impairment of frontocortical endocannabinoid degradation and high alcohol preference [online]Accessed 28 December 2005 URL: http://www.acnp.org/citations/NPP122805050541/default.pdf
  • HineBTorrelioMGershonS1975Attenuation of precipitated abstinence in methadone-dependent rats by delta 9-THCPsychopharmacol Commun1275831241452
  • HouchiHBabovicDPierreficheO2005CB1 receptor knockout mice display reduced ethanol-induced conditioned place preference and increased striatal dopamine D2 receptorsNeuropsychopharmacology303394915383833
  • HymanSEMalenkaRC2001Addiction and the brain: the neurobiology of compulsion and its persistenceNat Rev Neurosci269570311584307
  • JustinovaZTandaGMunzarP2004The opioid antagonist naltrexone reduces the reinforcing effects of Delta 9 tetrahydrocannabinol (THC) in squirrel monkeysPsychopharmacology1731869414668977
  • KaymakcalanSAyhanIHTulunayFC1977Naloxone-induced or post-withdrawal abstinence signs in delta9-tetrahydrocannabinol-tolerant ratsPsychopharmacology552439414285
  • KauerJA2004Learning mechanisms in addiction: synaptic plasticity in the ventral tegmental area as a result of exposure to drugs of abuseAnnu Rev Physiol664477514977410
  • KoobGFLe MoalM2001Drug addiction, dysregulation of reward, and allostasisNeuropsychopharmacology249712911120394
  • KoobGF1992Drugs of abuse: anatomy, pharmacology and function of reward pathwaysTrends Pharmacol Sci13177841604710
  • KoobGFAhmedSHBoutrelBChenSAKennyPJMarkouAO’DellLEParsonsLHSannaPP2004Neurobiological mechanisms in the transition from drug use to drug dependenceNeurosci Biobehav Rev277394915019424
  • KurtzDLStewartRBZweifelM1996Genetic differences in tolerance and sensitization to the sedative/hypnotic effects of alcoholPharmacol Biochem Behav53585918866959
  • LallemandFSoubriePHDe WittePH2001Effects of CB1 cannabinoid receptor blockade on ethanol preference after chronic ethanol administrationAlcohol Clin Exp Res2513172311584151
  • Le FollBGoldbergSR2004Rimonabant, a CB1 antagonist, blocks nicotine-conditioned place preferencesNeuroreport1521394315486497
  • LedentCValverdeOCossuG1999Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout miceScience28340149888857
  • LesscherHMHoogveldEBurbachJP2005Endogenous cannabinoids are not involved in cocaine reinforcement and development of cocaine-induced behavioural sensitizationEur Neuropsychopharmacol1531715572271
  • LichtmanAHSheikhSMLohHH2001Opioid and cannabinoid modulation of precipitated withdrawal in delta(9)-tetrahydrocannabinol and morphine-dependent miceJ Pharmacol Exp Ther29810071411504797
  • LupicaCRRiegelACHoffmanAF2004Marijuana and cannabinoid regulation of brain reward circuitsBr J Pharmacol1432273415313883
  • LupicaCRRiegelAC2005Endocannabinoid release from midbrain dopamine neurons: a potential substrate for cannabinoid receptor antagonist treatment of addictionNeuropharmacology4811051615878779
  • MaldonadoR2002Study of cannabinoid dependence in animalsPharmacol Ther951536412182962
  • MaldonadoRValverdeOBerrenderoF2006Involvement of the endocannabinoid system in drug addictionTrends Neurosci292253216483675
  • ManzanedoCAguilarMARodriguez-AriasM2004Cannabinoid agonist-induced sensitisation to morphine place preference in miceNeuroreport151373715167569
  • MartinMLedentCParmentierM2000Cocaine, but not morphine, induces conditioned place preference and sensitization to locomotor responses in CB1 knockout miceEur J Neurosci1240384611069600
  • Mas-NietoMPommierBTzavaraET2001Reduction of opioid dependence by the CB(1) antagonist SR141716A in mice: evaluation of the interest in pharmacotherapy of opioid addictionBr J Pharmacol13218091611309253
  • NaassilaMPierreficheOLedentC2004Decreased alcohol self-administration and increased alcohol sensitivity and withdrawal in CB1 receptor knockout miceNeuropharmacology462435314680762
  • NavarroMChowenJRocioA1998CB1 cannabinoid receptor antagonist-induced opiate withdrawal in morphine-dependent ratsNeuroreport933974029855288
  • NavarroMCarreraMRFrattaW2001Functional interaction between opioid and cannabinoid receptors in drug self-administrationJ Neurosci2153445011438610
  • NavarroMCarreraMRDel ArcoI2004Cannabinoid receptor antagonist reduces heroin self-administration only in dependent ratsEur J Pharmacol501235715464083
  • NestlerEJ2004Molecular mechanisms of drug addictionNeuropharmacology47Suppl 1243215464123
  • OrtizSOlivaJMPerez-RialS2004Differences in basal cannabinoid CB1 receptor function in selective brain areas and vulnerability to voluntary alcohol consumption in Fawn Hooded and Wistar ratsAlcohol Alcohol3929730215208160
  • ParolaroDViganoDRubinoT2005Endocannabinoids and drug dependenceCurr Drug Targets CNS Neurol Disord46435516375682
  • PierceRCKumaresanV2006The mesolimbic dopamine system: the final common pathway for the reinforcing effect of drugs of abuse?Neurosci Biobehav Rev302153816099045
  • RiceOVGordonNGiffordAN2002Conditioned place preference to morphine in cannabinoid CB1 receptor knockout miceBrain Res945135812113961
  • RomeroJFernandez-RuizJJVelaG1998Autoradiographic analysis of cannabinoid receptor binding and cannabinoid agonist-stimulated [35S]GTP gamma S binding in morphine-dependent miceDrug Alcohol Depend5024199649978
  • RubinoTMassiPViganoD2000Long-term treatment with SR141716A, the CB1 receptor antagonist, influences morphine withdrawal syndromeLife Sci6622131910834304
  • SchuckitMA1994Low level of response to alcohol as a predictor of future alcoholismAm J Psychiatry15118498296886
  • SerraSBrunettiGPaniM2002Blockade by the cannabinoid CB(1) receptor antagonist, SR 141716, of alcohol deprivation effect in alcohol-preferring ratsEur J Pharmacol44395712044797
  • SerraSCaraiMABrunettiG2001The cannabinoid receptor antagonist SR141716 prevents acquisition of drinking behavior in alcohol-preferring ratsEur J Pharmacol4303697111711056
  • SolinasMPanlilioLVAntoniouK2003The cannabinoid CB1 antagonist N-piperidinyl-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methylpyrazole-3-carboxamide (SR-141716A) differentially alters the reinforcing effects of heroin under continuous reinforcement, fixed ratio, and progressive ratio schedules of drug self-administration in ratsJ Pharmacol Exp Ther3069310212660305
  • SolinasMPanlilioLVTandaG2005Cannabinoid agonists but not inhibitors of endogenous cannabinoid transport or metabolism enhance the reinforcing efficacy of heroin in ratsNeuropsychopharmacology3020465715870833
  • SoriaGMendizabalVTourinoC2005Lack of CB1 cannabinoid receptor impairs cocaine self-administrationNeuropsychopharmacology3016708015742004
  • SpanoMSFattoreLCossuG2004CB1 receptor agonist and heroin, but not cocaine, reinstate cannabinoid-seeking behaviour in the ratBr J Pharmacol1433435015339858
  • TandaGPontieriFEDi ChiaraG1997Cannabinoid and heroin activation of mesolimbic dopamine transmission by a common mu1 opioid receptor mechanismScience2762048509197269
  • ValjentEMitchellJMBessonMJ2002Behavioural and biochemical evidence for interactions between Delta 9-tetrahydrocannabinol and nicotineBr J Pharmacol1355647811815392
  • ValverdeOMaldonadoRValjentE2000Cannabinoid withdrawal syndrome is reduced in pre-proenkephalin knock-out miceJ Neurosci209284911125007
  • Van SickleMDDuncanMKingsleyPJMouihateAUrbaniPMackieKStellaNMakriyannisAPiomelliDDavisonJSMarnettLJDi MarzoVPittmanQJPatelKDSharkeyKA2005Identification and functional characterization of brainstem cannabinoid CB2 receptorsScience3103293216224028
  • VelaGRuiz-GayoMFuentesJA1995Anandamide decreases naloxone-precipitated withdrawal signs in mice chronically treated with morphineNeuropharmacology3466587566503
  • ViganoDRubinoTParolaroD2005Molecular and cellular basis of cannabinoid and opioid interactionsPharmacol Biochem Behav81360815927245
  • WangLLiuJHarvey-WhiteJ2003Endocannabinoid signaling via cannabinoid receptor 1 is involved in ethanol preference and its age-dependent decline in miceProc Natl Acad Sci USA1001393812538878
  • WengerTMoldrichGFurstS2003Neuromorphological background of cannabis addictionBrain Res Bull61125812831997
  • XiZXGilbertJGPengXQ2006Cannabinoid CB1 receptor antagonist AM251 inhibits cocaine-primed relapse in rats: role of glutamate in the nucleus accumbensJ Neurosci268531616914679
  • YamaguchiTHagiwaraYTanakaH2001Endogenous cannabinoid, 2-arachidonoylglycerol, attenuates naloxone-precipitated withdrawal signs in morphine-dependent miceBrain Res909121611478928

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.