Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Drug and Alcohol Dependence 117 (2011) 102–110

Contents lists available at ScienceDirect

Drug and Alcohol Dependence


journal homepage: www.elsevier.com/locate/drugalcdep

Review

Involvement of the endocannabinoid system in alcohol dependence:


The biochemical, behavioral and genetic evidence
Amaia M. Erdozain a,b , Luis F. Callado a,b,∗
a
Department of Pharmacology, University of the Basque Country, 48940 Leioa, Bizkaia, Spain
b
Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain

a r t i c l e i n f o a b s t r a c t

Article history: Background: Recent advances in the understanding of alcohol dependence suggest that the endocannabi-
Received 19 October 2010 noid system (ECS) plays a key role in the neurobiological mechanisms underlying this pathology.
Received in revised form 7 February 2011 Methods: The aim of the present review is to show the currently available biochemical, behavioral and
Accepted 14 February 2011
genetic evidence on the involvement of the ECS in alcohol dependence.
Available online 16 March 2011
Discussion: Firstly, biochemical studies have shown that both chronic and acute administration of ethanol
produce alterations in different elements of this neurotransmission system. Secondly, the pharmacologi-
Keywords:
cal and genetic manipulation of the ECS in rodents result in altered ethanol-related behavior. Furthermore,
Alcohol dependence
Cannabinoids
rodent strains with different preference for ethanol differ in their ECS state. Also, genetic studies have
Endocannabinoid system described that particular polymorphisms in the genes coding for some elements of this system are associ-
Receptors ated with some phenotypes of alcohol dependence. Finally, the possible efficacy of cannabinoid receptor
Rimonabant blockers in the prevention of relapse to alcohol has been tested in clinical trials.
Conclusion: Altogether, these multiple lines of evidence suggest that the ECS is implicated in the devel-
opment of alcohol abuse and dependence.
© 2011 Elsevier Ireland Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
1.1. The endocannabinoid system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103
1.2. The endocannabinoid system and addiction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103
2. Biochemical evidence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103
3. Behavioral evidence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
3.1. Pharmacological modulation of the endocannabinoid system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
3.2. Genetic modulation of the endocannabinoid system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106
4. Genetic evidence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106
5. Rimonabant for the treatment of alcohol dependence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
Role of funding source . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108

1. Introduction morbidity, mortality and disability is very important. According to


the World Health Organization in 2000 alcohol dependence was
Alcohol dependence is a costly and socially devastating illness. responsible for 4% of global disease, constituting only a slightly
Ethanol produces a complex and multidimensional effect on health lower level than that caused by smoking (4.1%) and hypertension
and the global burden related to alcohol consumption in terms of (4.4%) (World Health Organization, 2004).
Ethanol is toxic to most body tissues, producing changes on
the cardiovascular system, digestive system, central nervous sys-
∗ Corresponding author at: Department of Pharmacology, University of the Basque tem, peripheral nerves, muscle–skeletal system and the fetus. In
Country, Leioa, Bizkaia, Spain. Tel.: +34 94 6012762; fax: +34 94 6013220. the central nervous system ethanol generates a deep depression of
E-mail address: lf.callado@ehu.es (L.F. Callado). neuronal functions, concomitantly decreasing glucose metabolism

0376-8716/$ – see front matter © 2011 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.drugalcdep.2011.02.003
A.M. Erdozain, L.F. Callado / Drug and Alcohol Dependence 117 (2011) 102–110 103

throughout the human brain (Wang et al., 2000). Nowadays, it is Matsuda et al., 1993), this regulatory mechanism is likely to be
well known that ethanol affects most of the neurochemical and indirect.
endocrine systems (Diamond and Gordon, 1997), among which is Additional data supporting the involvement of the ECS in the
the endocannabinoid system (ECS). reward pathway arises from in vivo microdialysis studies. Firstly,
The aim of the present review is to provide the most recent sci- acute alcohol, and nicotine, induced increase of dopamine levels in
entific evidence about the involvement of the ECS in the process the nucleus accumbens is significantly reduced by the CB1 receptor
of the development, tolerance, withdrawal, craving and relapse of antagonist rimonabant (Cohen et al., 2002). Similarly, in CB1 recep-
alcohol dependence. For this purpose we have performed a com- tor knockout mice acute ethanol administration did not produce
puterized search of the literature on PubMed and Scopus in order any dopamine release in the same region (Hungund et al., 2003).
to obtain biochemical, behavioral or genetic evidence. A secondary These studies confirm that the dopamine release induced by some
search was done by hand searching the reference lists from primary drugs of abuse is CB1 receptor dependent.
studies and former reviews. Nonetheless, the ECS is not only involved in the rewarding
properties of alcohol, but also in the motivation for drug seek-
1.1. The endocannabinoid system ing, the individual vulnerability for developing a dependence, the
acquisition of ethanol self-administering behavior, the develop-
The ECS constitutes a newly discovered system of neuromodu- ment of the withdrawal syndrome, etc. There is strong evidence
lation and comprises the cannabinoid receptors, along with their of the involvement of the ECS in alcohol dependence. In this
endogenous ligands, enzymes involved in the biosynthesis and article biochemical, behavioral and genetic evidence of this impli-
degradation of these ligands and putative membrane transport cation will be reviewed. Firstly, biochemical studies have shown
proteins. So far, two cannabinoid receptors have been identified that both chronic and acute administration of ethanol produce
and cloned. The CB1 receptor (Matsuda et al., 1990) is mainly alterations in different elements of the endocannabinoid system.
expressed in the brain, where it plays an important role in the Secondly, the pharmacological and genetic manipulation of this
regulation of neurotransmitter release (Wilson and Nicoll, 2002), system in rodents result in altered ethanol-related behavior. Fur-
and it is, indeed, the most abundant G-protein-coupled receptor ther, rodent strains with different preference for ethanol differ in
in the mammalian brain. Conversely, the CB2 receptor (Munro their ECS state. Finally, genetic studies have described that partic-
et al., 1993) is mainly located in peripheral tissues, at the high- ular polymorphisms in the genes coding for some elements of this
est levels in the immune system, such as spleen, tonsil, thymus neurotransmission system are associated with some phenotypes of
and lymphoid tissues (Galiegue et al., 1995). The endocannabinoids alcohol dependence.
are polyinsaturated fatty acids derived compounds, which share
some pharmacologic properties of 9 -THC. To date, anandamide 2. Biochemical evidence
(N-araquidonoylethanolamine, AEA) and 2-arachidonoylglycerol
(2-AG) are the most thoroughly studied endocannabinoids, which Several studies have demonstrated that chronic exposure to
are produced on demand and released in the extracellular environ- ethanol modulates some elements of the ECS both in vitro and in
ment, where they can bind and activate CB1 and CB2 cannabinoid vivo (Table 1). The first in vitro study reported a concentration- and
receptors among others. Their hydrolysis, resulting in the termina- time-dependent increase in anandamide formation after chronic
tion of endocannabinoid signaling, is an intracellular event carried exposure of neuroblastoma SK-N-SH strain cells to ethanol. This
out by different enzymes, mainly the fatty acid amide hydrolase increase in the endocannabinoid formation was inhibited by the
(FAAH) and monoacylglycerol lipase (Alexander and Kendall, 2009). CB1 receptor antagonist rimonabant and pertussis toxin, sug-
FAAH is the main enzyme involved in AEA degradation, produc- gesting a CB1 receptor and Gi/o protein mediated regulation
ing arachidonic acid and ethanolamime. In neural tissues, FAAH (Basavarajappa and Hungund, 1999). Shortly thereafter, the stim-
immunoreactivity is associated primarily with neurons, in a pat- ulation of the other main endocannabinoid formation, the 2-AG,
tern extensively complementary to the expression of CB1 receptors was reported after chronic exposure of cerebellar granule neuron
(Tsou et al., 1998). cultures to ethanol, being similarly inhibited by rimonabant and
pertussis toxin (Basavarajappa et al., 2000).
1.2. The endocannabinoid system and addiction The modulation of the endocannabinoid content by chronic
ethanol has also been observed in rodent brain, although the results
There is much evidence suggesting an association between the are not consistent among all the studies. Thus, anandamide lev-
neuroanatomical distribution of the ECS and some of the central els were increased in the limbic forebrain while decreased in the
effects produced by ethanol. More precisely, CB1 receptor localiza- midbrain of ethanol-treated rats (Gonzalez et al., 2002a, 2004).
tion in the cerebral cortex, hippocampus, thalamus, basal ganglia, Another study corroborated this increase of anandamide in mice
cerebellum and medulla is consistent with the effects of alcohol on cerebral cortex after chronic ethanol exposure, which was con-
higher cognitive and motor functions, memory, hypothermia and sistent with a concomitant significant reduction in FAAH activity
nociception (Herkenham et al., 1991; Glass et al., 1997). (Vinod et al., 2006). In regard to 2-AG, an in vivo microdialysis
The crucial role of the mesocorticolimbic dopaminergic path- study reported increased dialysate levels of this endocannabi-
way in the reward and reinforcement produced by all drugs of noid in the nucleus accumbens of ethanol self-administering rats,
abuse, including alcohol, has thoroughly been documented (Koob with no concomitant change in anandamide concentrations (Caille
and Volkow, 2010). In this way, the ECS seems to be involved et al., 2007). However, other studies observed either a decrease
in the regulation of this reward pathway. Thus, CB1 cannabinoid (Gonzalez et al., 2002a) or no change (Gonzalez et al., 2004) in
receptors are present in different regions of the mesocorticolim- 2-AG levels in rat midbrain. Nevertheless, these endocannabinoid
bic circuitry and are involved in the neurobiological mechanisms levels modulated by chronic ethanol administration are further
underlying the addictive processes of drugs of abuse (Maldonado affected by alcohol-deprivation and relapse to alcohol consumption
et al., 2006). It has been described that following depolarization (Gonzalez et al., 2004).
dopaminergic neurons in the ventral tegmental area (VTA) release Cannabinoid CB1 receptor mRNA, density and functionality have
endocannabinoids that act as a regulatory feedback mechanism also been reported to be altered by chronic exposure to ethanol. The
(Lupica and Riegel, 2005). However, since dopaminergic neurons first study indicated a significant down-regulation of the CB1 recep-
do not appear to express CB1 receptor (Herkenham et al., 1991; tor density in whole mouse brain chronically exposed to ethanol,
104 A.M. Erdozain, L.F. Callado / Drug and Alcohol Dependence 117 (2011) 102–110

Table 1
Biochemical evidence about the involvement of the endocannabinoid system in alcohol dependence.

Type of study Results Reference

In vitro studies
Chronic ethanol increased AEA formation in SK-N-SH cells Basavarajappa and Hungund (1999)
ECBs
Chronic ethanol increased 2-AG formation in cerebellar granule neurons Basavarajappa et al. (2000)

Animal brain studies


Chronic ethanol decreased AEA and 2-AG contents in the midbrain and increased AEA content Gonzalez et al. (2002a)
ECBs in the limbic forebrain
Levels of ECBs underwent significant changes in reward-related rat brain areas during Gonzalez et al. (2004)
alcoholization, deprivation and relapse
Ethanol self- administration increased 2-AG levels, without altering AEA levels, in rat nucleus Caille et al. (2007)
accumbens (in vivo microdialysis)
Chronic ethanol down-regulated CB1Rs in whole mouse brain membranes Basavarajappa et al. (1998)
Chronic ethanol was usually ineffective in altering CB1R binding and mRNA levels in all rat Gonzalez et al. (2002b)
CB1R
brain regions examined
Chronic ethanol reduced CB1R mRNA expression in some rat brain regions Ortiz et al. (2004a)
Ethanol desensitized CB1R modulating the synthesis of monoamines Moranta et al. (2006)

Chronic ethanol reduced CB1 receptor binding and functionality in several mouse brain Vinod et al. (2006)
CB1R & ECBs
regions; increased AEA levels and reduced FAAH activity in cortex
Chronic intermittent ethanol withdrawal transiently down-regulated rat hippocampal CB1Rs, Mitrirattanakul et al. (2007)
followed by long-term up-regulation, with increased EBCs
Human brain studies
CB1R & ECBs Elevated CB1R density and functionality and ECBs in the prefrontal cortex of alcoholic suicide Vinod et al. (2005)
victims compared to non-suicidal alcoholics
CB1R & FAAH Decreased CB1R density and functionality and FAAH activity in the ventral striatum of Vinod et al. (2010)
non-suicidal alcoholics compared to controls
ECBs Decreased AEA in nucleus accumbens and frontal cortex of Cloninger type 1 alcoholics Lehtonen et al. (2010)

Acute alcohol
Reduced ECB levels in different rat brain regions Rubio et al. (2007)
ECBs Reduced AEA in different rat brain regions, which was not dependent on FAAH or NAT activity Ferrer et al. (2007)
Reduced AEA, increased 2-AG in rat nucleus accumbens (in vivo microdialysis) Alvarez-Jaimes et al. (2009)

CB1R Reduced CB1R mRNA in different rat brain regions Oliva et al. (2008)
CB1R & FAAH Reduced CB1R levels in the rat amygdala and prefrontal cortex; FAAH activity was not Rubio et al. (2009)
increased in any of the regions analyzed

ECBs: endocannabinoid levels. CB1R: CB1 receptor.

without any changes in the affinity (Basavarajappa et al., 1998). hol dependence. A hyperactivity of the endocannabinoid signaling
This finding agrees with a reduction in the CB1 receptor density and has been reported in the prefrontal cortex of suicidal alcoholic sub-
functionality observed in particular brain regions of mice follow- jects compared to alcoholic subjects dying of other causes than
ing the same ethanol exposure (Vinod et al., 2006). Similarly, CB1 suicide. These suicidal alcoholic subjects presented higher CB1
receptor mRNA expression was found to be decreased in several receptor density and functionality, as well as higher anandamide
rat brain regions after chronic ethanol consumption (Ortiz et al., and 2-AG levels. The authors suggested that this endocannabinoid
2004a). Nonetheless, another study did not observe any changes hyperactivity in the prefrontal cortex could be a potential factor
after chronic exposure to ethanol either in CB1 receptor binding for suicide in alcohol dependence (Vinod et al., 2005). Recently,
or mRNA levels in rat brain (Gonzalez et al., 2002b). In regard to the same group has described decreased CB1 receptor binding
the neuromodulation that exerts the CB1 receptor over neurotrans- and functionality in the ventral striatum of non-suicidal alcoholic
mitter synthesis and release, ethanol also produces a functional subjects compared to controls. However, these parameters were
desensitization of CB1 receptors in rats, modulating the synthesis elevated in the suicidal alcoholics when compared to non-suicidal
of brain monoamines (Moranta et al., 2006). alcoholic subjects (Vinod et al., 2010). In regard to the differences
Some studies have focused on the effect of alcohol with- between Cloninger type 1 and 2 alcoholics, decreased anandamide
drawal on the CB1 receptors. A transient down-regulation of levels were observed in the nucleus accumbens and frontal cortex
these receptors followed by a long-term up-regulation, with con- in type 1 alcoholics (Lehtonen et al., 2010). Thus, studies performed
comitant increased endocannabinoid levels, has been described in in human brain show contradictory results probably due to con-
rat hippocampus after chronic intermittent ethanol withdrawal founding variables as suicide.
(Mitrirattanakul et al., 2007). Similarly, a recovery of the decreased Several recent reports have shown that acute alcohol adminis-
CB1 receptor binding and functionality has also been reported after tration does also induce changes in the endocannabinoid system.
withdrawal (Vinod et al., 2006). Reduced levels of endocannabinoids were observed in different
On the whole, the biochemical studies performed in rodents that brain regions after short-term exposure to ethanol (Rubio et al.,
chronically consume ethanol suggest a down-regulation of brain 2007). Similarly anandamide content was also found reduced in
CB1 receptors, which is likely to be the consequence of high endo- several rat brain regions, which was not dependent on FAAH or N-
cannabinoid levels observed in these animals. The reason for this acyltransferase activity (Ferrer et al., 2007). An in vivo microdialysis
high endocannabinoid content is still unknown and might be either study confirmed this reduction of dyalisate anandamide levels in
a higher synthesis rate, a lower degradation rate or even a decreased the nucleus accumbens after an acute ethanol injection, but con-
re-uptake of the synaptic endocannabinoids. versely observed an increase in 2-AG levels (Alvarez-Jaimes et al.,
In recent years some studies have been performed in postmortem 2009). Furthermore, CB1 receptors are also affected by short-term
human brain of subjects that had previously been diagnosed of alco- exposure to ethanol, since a decrease in the receptor mRNA and pro-
A.M. Erdozain, L.F. Callado / Drug and Alcohol Dependence 117 (2011) 102–110 105

Table 2
Behavioral evidence about the involvement of the endocannabinoid system in alcohol dependence, after pharmacological manipulation of the system.

Modulation Results Reference

Agonists
CP55,940 Increased the motivation for consuming beer in rats Gallate et al. (1999)
WIN 55,212-2 and CP55,940 Increased ethanol intake in alcohol preferring Sardinian rats Colombo et al. (2002)
WIN 55,212-2 Increased alcohol relapse during a period of alcohol deprivation Alen et al. (2008)

Antagonists
Reduced ethanol intake in alcohol-preferring mice Arnone et al. (1997)
Reduced ethanol intake in alcohol-preferring rats Colombo et al. (1998) and Dyr et al. (2008)
Reduced the motivation for consuming beer in rats Gallate and McGregor (1999)
Reduced ethanol self-administration in alcohol dependent rats Rodriguez de Fonseca et al. (1999)
Reduced ethanol preference after chronic alcoholization in rats Lallemand et al. (2001)
Rimonabant (SR141716) Reduced ethanol seeking and consumption in rats Freedland et al. (2001)
Prevented acquisition of drinking behavior in alcohol-preferring rats Serra et al. (2001)
Abolished the alcohol deprivation effect in alcohol-preferring rats Serra et al. (2002)
Abolished motivational properties of alcohol in alcohol-preferring rats Colombo et al. (2004)
Reduced ethanol self-administration and conditioned reinstatement of Cippitelli et al. (2005)
ethanol-seeking in both alcohol-preferring and non-preferring rats
Reduced ethanol self-administration in alcohol-preferring rats when Hansson et al. (2007)
administered systemically or locally into the prefrontal cortex
Reduced ethanol intake in mice and rats Rinaldi-Carmona et al. (2004)
Surinabant (SR147778) Reduced ethanol intake and motivational properties in alcohol-preferring rats Gessa et al. (2005)
Reduced ethanol preference after chronic alcoholization in rats Lallemand and De Witte (2006)

SLV330 Reduced ethanol self-administration and reinstatement of ethanol seeking in De Bruin et al. (2011)
rats
FAAH inhibitors
Increased ethanol self-administration in rats Hansson et al. (2007)
URB597 Increased ethanol preference and consumption in mice Blednov et al. (2007)
Increased ethanol preference in mice Vinod et al. (2008)

Anandamide transporter inhibitors


AM404 Reduced ethanol self-administration in rats Cippitelli et al. (2007)

tein levels has been reported in particular rat brain regions (Oliva or not to pulmonary chronic alcoholization (Lallemand et al., 2001).
et al., 2008; Rubio et al., 2009). In other type of studies animals are required to complete an oper-
In summary, chronic or acute alcohol administration seem to ant response in order to obtain access to alcohol, which allows
produce different effects in the ECS. These differences may be due studying the motivation of the animal to consume alcohol. Thus,
to the adaptative responses produced by the chronic exposure to rimonabant reduced motivation for alcohol in a dose-dependent
ethanol. manner both in non-preferring (Gallate and McGregor, 1999) and
preferring rats (Colombo et al., 2004). Similarly, operant ethanol
3. Behavioral evidence self-administration was reduced by rimonabant in rats tested under
different procedures (Freedland et al., 2001; Rodriguez de Fonseca
3.1. Pharmacological modulation of the endocannabinoid system et al., 1999; Caille et al., 2007; Cippitelli et al., 2005; Hansson et al.,
2007). Further, rimonabant prevented the acquisition of ethanol
A wide variety of reports have demonstrated that the pharmaco- drinking behavior in alcohol-preferring rats (Serra et al., 2001).
logical modulation of the endocannabinoid system, by cannabinoid In order to study the withdrawal and relapse in animals, two
agonists, antagonists, FAAH inhibitors or anandamide transporter experimental procedures have been developed. Firstly, the alcohol
inhibitors, alters the ethanol-related behavior in rodents (summa- deprivation effect consists in the temporary increase in volun-
rized in Table 2). These findings further support the involvement of tary ethanol intake after a period of alcohol withdrawal. Indeed,
the ECS in neurobiology underlying alcohol dependence. rimonabant completely abolished the alcohol deprivation effect
First of all, cannabinoid agonists WIN 55,212-2 and CP 55,940, in alcohol-preferring rats (Serra et al., 2002). The second pro-
have been shown to increase ethanol consumption and prefer- cedure assesses the reinstatement of alcohol-seeking behavior
ence. They produced an augment in the motivation for beer in rats induced by a specific stimulus which was previously associated
(Gallate et al., 1999), in addition to enhance ethanol intake in mice with alcohol availability. In the same way, rimonabant reduced
(Kelai et al., 2006) and in a selectively bred alcohol-preferring Sar- conditioned reinstatement of ethanol-seeking behavior both in
dinian rat strain (Colombo et al., 2002). Furthermore, WIN 55,212-2 alcohol-preferring and non-preferring rats (Cippitelli et al., 2005).
increased alcohol relapse during a period of alcohol deprivation in Similar results to those obtained with rimonabant have
rats (Alen et al., 2008). been further observed with another cannabinoid antagonist that
Conversely, the administration of the cannabinoid antago- was more recently synthesized, the surinabant or SR1417778
nists rimonabant (SR141716) and surinabant (SR147778) reduces (Rinaldi-Carmona et al., 2004). A first study indicated that under
alcohol-related behavior and consumption. The first research work the two-bottled choice paradigm surinabant dose-dependently
to study this issue showed that rimonabant inhibited ethanol intake decreased ethanol consumption in mice (Rinaldi-Carmona et al.,
in selectively bred alcohol-preferring mice tested under the two- 2004). Likewise, surinabant reduced ethanol intake and preference
bottle choice paradigm (Arnone et al., 1997). This phenomenon in alcohol-preferring rats (Gessa et al., 2005) and non-preferring
was later confirmed in different strains of alcohol-preferring rats rats that had been chronically alcoholized (Lallemand and De Witte,
(Colombo et al., 1998; Dyr et al., 2008), as well as in non-preferring 2006). Also, this cannabinoid antagonist dose-dependently reduced
mice (Poncelet et al., 2003) and both non-preferring rats exposed the acquisition of ethanol drinking in alcohol-preferring rats and
106 A.M. Erdozain, L.F. Callado / Drug and Alcohol Dependence 117 (2011) 102–110

Table 3
Behavioral evidence about the involvement of the endocannabinoid system in alcohol dependence, after genetic manipulation of the system.

Modulation Results Reference

CB1R knockout mice


Decreased ethanol intake and absence of alcohol-induced DA release in N accumbens Hungund et al. (2003)
Absence of ethanol withdrawal symptoms and stress-stimulated ethanol drinking Racz et al. (2003)
Decreased ethanol intake and preference in young mice Wang et al. (2003)
Decreased ethanol intake Poncelet et al. (2003)
Decreased ethanol self-administration and increased alcohol sensitivity and withdrawal Naassila et al. (2004)
Decreased ethanol-induced conditioned place preference Houchi et al. (2005)
Decreased ethanol preference; increased blood ethanol concentration at high ethanol doses Lallemand and De Witte (2005)
Decreased ethanol intake and preference; absence of conditioned place preference Thanos et al. (2005)

FAAH knockout mice


Increased ethanol intake and preference; decreased sensitivity in female FAAH−/− mice Basavarajappa et al. (2006)
Increased ethanol intake and preference; no differences in the ethanol-induced place preference, sensitivity or withdrawal Blednov et al. (2007)
Increased ethanol preference; decreased ethanol sensitivity and withdrawal convulsions Vinod et al. (2008)

suppressed the alcohol deprivation effect after a period of alcohol Secondly, the ethanol-related behavior has also been studied
abstinence (Gessa et al., 2005). in FAAH knockout mice, which indeed have higher levels of endo-
Recently, it has been reported that another CB1 cannabinoid cannabinoids compared to their wild type mates. All the studies
receptor antagonist, SLV330, was effective in reducing ethanol self- agree that these mice show increased intake and preference for
administration and reinstatement of ethanol seeking in rats (De ethanol (Basavarajappa et al., 2006; Blednov et al., 2007; Vinod
Bruin et al., 2011). et al., 2008), although one of them only observed this phenomena
The indirect activation of the ECS has also been studied in regard in female mice, suggesting a sex-link mechanism (Basavarajappa
to alcohol dependence. Thus, FAAH inhibitor URB597 administra- et al., 2006). Also, FAAH knockout mice displayed lower sensitivity
tion increased operant ethanol self-administration in rats (Hansson to acute effects of ethanol, and a reduction in ethanol withdrawal
et al., 2007), and similarly produced an increase in ethanol prefer- convulsions (Vinod et al., 2008). However, Blednov et al. (2007) did
ence and consumption in mice in a two-bottle choice paradigm not observe any difference in ethanol sensitivity or withdrawal.
(Blednov et al., 2007; Vinod et al., 2008). On the other hand, the The findings of this section show that the stimulation of the ECS
acute administration of AM404, an inhibitor of the putative anan- increases ethanol consumption, whereas a blockade of the system
damide transporter, reduced ethanol self-administration in rats reduces ethanol administration. These effects may be due to a possi-
(Cippitelli et al., 2007). ble mechanism through which the endocannabinoids may enhance
the reinforcing properties of ethanol.
3.2. Genetic modulation of the endocannabinoid system
4. Genetic evidence
An additional approach to understand the involvement of the
ECS in particular situations relies on the genetic modulation of the Alcohol dependence is a heterogeneous complex disorder with a
system. So far, CB1 receptor and fatty acid amide hydrolase (FAAH) multiple genetic background. Twin studies on alcohol dependence,
enzyme knockout mice have been studied (summarized in Table 3) one of the classical genetic approaches, have revealed a heritabil-
and show altered ethanol-related behavior. ity of alcohol dependence of over 50% (between 40 and 70%)
First of all, mice lacking CB1 receptor exhibited markedly (Köhnke, 2008). Thus, vulnerability for excessive alcohol consump-
reduced voluntary alcohol consumption in a two-bottle choice tion or development of alcohol dependence has been associated
paradigm (Hungund et al., 2003; Poncelet et al., 2003), which was with genes involved in metabolism of ethanol and also several
concomitant with a complete lack of ethanol-induced dopamine genes of different neurotransmission systems that underlie alcohol
release in the nucleus accumbens (Hungund et al., 2003). A similar dependence.
study observed this decrease in ethanol intake, but only in young In this context, genetic evidence about the involvement of
mice that did not express CB1 receptors, since old mice showed the ECS in alcohol dependence has risen from the study of
similar preference for ethanol regardless of their genetic back- rodent strains that differ in their preference for ethanol (summa-
ground (Wang et al., 2003). The reduction in ethanol consumption rized in Table 4). First of all, differences in CB1 receptor density
was further confirmed in an operant self-administration paradigm and functionality have been reported within animals with dif-
(Naassila et al., 2004; Thanos et al., 2005). Also, these animals show ferent preference for ethanol. On one hand, alcohol-preferring
a reduced ethanol-induced conditioned place preference, which mice showed decreased CB1 receptor density, but higher affin-
further suggests the involvement of the ECS in alcohol depen- ity and functionality than alcohol-avoiding mice (Hungund and
dence, beyond the rewarding properties of ethanol (Houchi et al., Basavarajappa, 2000; Basavarajappa and Hungund, 2001). On the
2005; Thanos et al., 2005). Furthermore, ethanol withdrawal is sig- other hand, both decreased CB1 receptor gene expression and
nificantly altered in CB1 receptor knockout mice, although there functionality have been observed in different brain areas of alcohol-
are discrepancies in the results. Racz et al. (2003) reported that preferring rats (Ortiz et al., 2004b). Moreover, in the prefrontal
ethanol withdrawal symptoms were completely absent in these cortex of alcohol-preferring rats, not only has a decrease in CB1
genetically modified mice, while Naassila et al. (2004) observed an receptor binding and functionality been reported, but a decrease
increase in alcohol sensitivity and withdrawal induced convulsions. in FAAH gene expression and activity has also been described,
Additionally, ethanol induced higher blood ethanol concentrations together with an increase in 2-AG levels (Hansson et al., 2007).
in CB1 knockout mice, while decreasing ethanol preference. This Genetic studies in humans have also described that particu-
fact revealed that CB1 receptors might also be involved in ethanol lar polymorphisms in the genes coding for some elements of the
absorption and distribution, particularly after administration of ECS could be associated with some phenotypes of alcohol depen-
high ethanol doses (Lallemand and De Witte, 2005). dence (Table 4). However, although a great number of genetic
A.M. Erdozain, L.F. Callado / Drug and Alcohol Dependence 117 (2011) 102–110 107

Table 4
Genetic evidence about the involvement of the endocannabinoid system in drug addiction and alcohol dependence.

Study Results Reference

Rodent strains with different alcohol preference


Decreased CB1R density but higher affinity in alcohol-preferring mice Hungund and Basavarajappa (2000)
CB1R Increased CB1R functionality in alcohol-preferring mice Basavarajappa and Hungund (2001)
Decreased CB1R mRNA and functionality in alcohol-preferring rats Ortiz et al. (2004b)
CB1R, FAAH & ECBs Decreased CB1R binding and functionality; decreased FAAH expression and Hansson et al. (2007)
activity; increased 2-AG, in the prefrontal cortex of alcohol-preferring rats
CB1R polymorphisms
Association with severe alcohol withdrawal syndrome Schmidt et al. (2002)
rs1049353 No significant association with different alcoholism-related phenotypes, Preuss et al. (2003)
including severe alcohol withdrawal syndrome
This meta-analysis showed no significant association with alcohol Benyamina et al. (2011)
dependence;
rs6454674 Association with alcohol dependence Zuo et al. (2007)
rs2023239 Association with CB1R density, alcohol-elicited brain activation, subjective Hutchison et al. (2008)
reward when consuming alcohol and positive outcomes with
pharmacotherapy that targets the dopaminergic system
Association with alcoholic patients who suffered from attention deficit/ Ponce et al. (2003)
AAT repeat
hyperactivity disorder during childhood
microsatellite
No significant association with alcohol dependence; although significant with Comings et al. (1997)
cocaine, amphetamine and cannabis dependence
Significant association with illicit substance dependence Benyamina et al. (2011)

No significant association of different SNPs with alcohol dependence in a Japanese population Herman et al. (2006)
Association of a three-SNP haplotype with alcohol dependence Zhang et al. (2004)

FAAH polymorphisms
Association with concomitant street drug and problem drug/alcohol use Sipe et al. (2002)
rs324420 Association with multiple different drug addictions Flanagan et al. (2006)
Association with reward-related ventral striatum reactivity Hariri et al. (2009)

CB2R polymorphisms
rs2501432 Association with alcoholism in a Japanese population Ishiguro et al. (2007)

MAGL polymorphisms
No significant association of any of the studied SNP of MAGL and FAAH with alcoholism in a Japanese population Iwasaki et al. (2007)

ECBs: endocannabinoid levels. CB1R: CB1 receptor.

studies have been performed, only a few have found a significant ing alcohol and more positive outcomes with pharmacother-
association. First of all, a particular single nucleotide polymor- apy that targets the dopaminergic mesocorticolimbic circuitry
phism (SNP) of the CB1 receptor, the rs1049353, was associated (Hutchison et al., 2008).
with severe alcohol withdrawal syndrome (Schmidt et al., 2002). A single nucleotide polymorphism in the FAAH gene, the
Nonetheless, another study that assessed the same particular rs324420, which produces a mutant enzyme with reduced cellu-
variation failed to confirm the association with different alcohol lar stability and therefore reduced expression and activity (Chiang
dependence-related phenotypes, including severe withdrawal syn- et al., 2004), has also been studied in regard to alcohol and other
drome (Preuss et al., 2003). Further, a nine-allele microsatellite drug use, with positive outcomes. The first study observed an asso-
polymorphism of the CB1 receptor, containing repeats of the single ciation of this SNP with concomitant street drug use and problem
trinucleotide AAT, has been associated with childhood attention drug/alcohol use (Sipe et al., 2002). Another study confirmed this
deficit/hyperactivity disorder in alcoholic patients (Ponce et al., association in a group composed of subjects with different drug
2003). For this microsatellite repeat, again, another study did not addictions, including alcohol dependence (Flanagan et al., 2006).
find an association with alcohol dependence, although it showed Recently, an imaging study has reported that the A carriers for
an association with other drug dependences and with intravenous this polymorphism have increased brain reactivity in the reward-
drug use (Comings et al., 1997). In the same context, a recent related ventral striatum, which highlights the importance of the
meta-analysis did not observe any significant association of either variability in the endocannabinoid signaling in behavioral pro-
rs1049353 or AAT repeat polymorphisms with alcohol dependence cesses related to addiction (Hariri et al., 2009).
(Benyamina et al., 2011). Similarly, another study failed to find an To conclude, two other genes of the ECS have been studied in
association of 4 different CB1 receptor SNPs with alcohol depen- alcohol dependence. A significant association of the CB2 recep-
dence in a Japanese population (Herman et al., 2006). tor polymorphism rs2501432 with alcohol dependence has been
Three additional studies have provided positive association of reported in Japanese patients (Ishiguro et al., 2007). Also, 14 differ-
CB1 receptor variants with alcohol dependence. Thus a highly sig- ent polymorphisms in the monoacylglycerol lipase (MAGL) gene
nificant difference was observed in a particular haplotype of three were studied in a Japanese alcoholic population, but none of the
different SNPs in Japanese alcoholics compared to controls (Zhang markers showed a significant association (Iwasaki et al., 2007).
et al., 2004). Also, risk for alcohol dependence was significantly Many of the studies presented show genetic evidence about the
associated with the rs6454674 single nucleotide polymorphism. involvement of the ECS in alcohol dependence. However, the lack
Furthermore, recently, the C allele for the rs2023239 CB1 recep- of positive associations in some studies can be attributed to the
tor SNP was associated with greater CB1 receptor density in the genetic differences between the populations of alcoholics and con-
prefrontal cortex of alcoholic patients, greater alcohol-elicited trols studied. Therefore, further studies are required to verify the
brain activation, greater subjective reward when consum- tentative allelic and genotypic associations reported.
108 A.M. Erdozain, L.F. Callado / Drug and Alcohol Dependence 117 (2011) 102–110

5. Rimonabant for the treatment of alcohol dependence cological treatments to decrease not only ethanol intake, but also
the motivation for consuming alcohol.
As reviewed throughout the article, a great amount of evidence
suggests the involvement of the ECS in the neurobiology of alcohol Role of funding source
dependence. More precisely, CB1 receptor blockade by rimonabant
in rodents has been reported to decrease not only ethanol intake Funding for this study was provided by Plan Nacional sobre Dro-
and preference, but also the motivation for consuming the drug, gas (PI 20061045), Gobierno Vasco (IT-199-07), and the Instituto de
along with the alcohol deprivation effect and the conditioned rein- Salud Carlos III, Centro de Investigación Biomédica en Red de Salud
statement of ethanol-seeking behavior. In this context, two Phase II Mental, CIBERSAM, Spain. AM Erdozain is recipient of a predoctoral
clinical studies have been conducted in human alcoholics in order fellowship from the Basque Government. The funding sources had
to study the possible utility of rimonabant in alcohol dependence. no further role in the writing of the review, or in the decision to
The first clinical trial was a 12-week double-blind, placebo- submit the paper for publication.
controlled study to evaluate the efficacy of rimonabant 20 mg/day
in the prevention of alcohol relapse in alcoholic patients that had
recently been detoxified (Soyka et al., 2008). Treatment compliance Contributors
tended to be better in the group of patients receiving rimonabant
than in the control group (patients receiving placebo). Overall, the AM Erdozain performed the literature search and LF Callado
clinical and biological safety and tolerability of rimonabant was wrote the first draft of the manuscript. Both authors contributed
good, and the rate of adverse effects was similar in both groups. The to and have approved the final manuscript.
primary end point of the study was to assess the time to first drink
and the time to relapse to first heavy alcohol drinking. At the end Conflict of interest
of the study there was a little favorable effect of rimonabant in pre-
venting relapse, although it did not reach statistical signification: There are no conflicts of interest.
41.5% vs 47.7% was the general relapse rate, and 27.7% vs 35.6% was
the relapse rate to heavy drinking. In regard to the secondary end
References
points of the study, there were no significant differences between
groups in cumulative abstinence duration in days, percentage of Alen, F., Moreno-Sanz, G., Isabel de Tena, A., Brooks, R.D., Lopez-Jimenez, A., Navarro,
drinking days and percentage of heavy drinking days, although M., Lopez-Moreno, J.A., 2008. Pharmacological activation of CB1 and D2 recep-
tors in rats: predominant role of CB1 in the increase of alcohol relapse. Eur. J.
there was similarly a trend toward favorable effects of rimona-
Neurosci. 27, 3292–3298.
bant. Authors suggest that the lack of improvement of rimonabant Alexander, S.P.H., Kendall, D.A., 2009. The life cycle of the endocannabinoids: for-
over placebo might be explained, firstly, by a very high benefi- mation and inactivation. In: Alexander, S.P.H., Kendall, D.A. (Eds.), Behavioral
cial response rate in the placebo group, and secondly, by the short Neurobiology of the Endocannabinoid System. Springer, New York.
Alvarez-Jaimes, L., Stouffer, D.G., Parsons, L.H., 2009. Chronic ethanol treatment
duration of the treatment. potentiates ethanol-induced increases in interstitial nucleus accumbens endo-
The second clinical trial was a 3-week double-blind, placebo- cannabinoid levels in rats. J. Neurochem. 111, 37–48.
controlled study to evaluate the effect of rimonabant 20 mg/day Arnone, M., Maruani, J., Chaperon, F., Thiebot, M.H., Poncelet, M., Soubrie, P., Le Fur,
G., 1997. Selective inhibition of sucrose and ethanol intake by SR 141716, an
on alcohol consumption in nontreatment-seeking heavy alcohol antagonist of central cannabinoid (CB1) receptors. Psychopharmacology (Berl.)
drinkers (George et al., 2010). First of all, after 1 week baseline, 18 132, 104–106.
participants received rimonabant and 21 received placebo during 2 Basavarajappa, B.S., Cooper, T.B., Hungund, B.L., 1998. Chronic ethanol adminis-
tration down-regulates cannabinoid receptors in mouse brain synaptic plasma
weeks, and they reported their daily alcohol consumption by tele- membrane. Brain Res. 793, 212–218.
phone. Rimonabant was not shown to alter alcohol consumption. Basavarajappa, B.S., Hungund, B.L., 1999. Chronic ethanol increases
Secondly, after the 2 weeks treatment, all patients were evaluated the cannabinoid receptor agonist anandamide and its precursor N-
arachidonoylphosphatidylethanolamine in SK-N-SH cells. J. Neurochem.
in an alcohol self-administration paradigm in which after receiving
72, 522–528.
an oral priming dose of ethanol they had the option of consuming Basavarajappa, B.S., Hungund, B.L., 2001. Cannabinoid receptor agonist-stimulated
up to eight alcohol drinks or receiving some money for each non- [35S]guanosine triphosphate gammaS binding in the brain of C57BL/6 and DBA/2
mice. J. Neurosci. Res. 64, 429–436.
consumed drink. The results show that rimonabant did not change
Basavarajappa, B.S., Saito, M., Cooper, T.B., Hungund, B.L., 2000. Stimulation of
alcohol self-administration either. cannabinoid receptor agonist 2-arachidonylglycerol by chronic ethanol and its
Rimonabant was commercialized for the treatment of obesity. modulation by specific neuromodulators in cerebellar granule neurons. Biochim.
Nevertheless, due to the incidence of severe adverse psychiatric Biophys. Acta 1535, 78–86.
Basavarajappa, B.S., Yalamanchili, R., Cravatt, B.F., Cooper, T.B., Hungund, B.L., 2006.
effects during the treatment (Christensen et al., 2007), its commer- Increased ethanol consumption and preference and decreased ethanol sensitiv-
cialization has been recently discontinued (European Medicines ity in female FAAH knockout mice. Neuropharmacology 50, 834–844.
Agency, 2009). Benyamina, A., Kebir, O., Blecha, L., Reynaud, M., Krebs, M.O., 2011. CNR1 gene poly-
morphisms in addictive disorders: a systematic review and a meta-analysis.
Addict. Biol. 16, 1–6.
Blednov, Y.A., Cravatt, B.F., Boehm, S.L., Walker, D., Harris, R.A., 2007. Role of endo-
6. Conclusion cannabinoids in alcohol consumption and intoxication: studies of mice lacking
fatty acid amide hydrolase. Neuropsychopharmacology 32, 1570–1582.
Caille, S., Alvarez-Jaimes, L., Polis, I., Stouffer, D.G., Parsons, L.H., 2007. Specific alter-
In conclusion, several lines of evidence support the involvement ations of extracellular endocannabinoid levels in the nucleus accumbens by
of the ECS in alcohol dependence. It is clear that this neurotransmis- ethanol, heroin, and cocaine self-administration. J. Neurosci. 27, 3695–3702.
Chiang, K.P., Gerber, A.L., Sipe, J.C., Cravatt, B.F., 2004. Reduced cellular expression
sion system is directly involved in the primary rewarding effects of
and activity of the P129T mutant of human fatty acid amide hydrolase: evidence
ethanol through different cellular mechanisms. Moreover, the ECS for a link between defects in the endocannabinoid system and problem drug use.
seems to play an important role in the motivation to seek the drug Hum. Mol. Genet. 13, 2113–2119.
Christensen, R., Kristensen, P.K., Bartels, E.M., Bliddal, H., Astrup, A., 2007. Efficacy
in alcoholics and in the relapse to drug-seeking in alcoholic patients
and safety of the weight-loss drug rimonabant: a meta-analysis of randomised
that had recently been detoxified. Further studies will be required trials. Lancet 370, 1706–1713.
to clarify the specific mechanisms that mediate these effects. How- Cippitelli, A., Bilbao, A., Gorriti, M.A., Navarro, M., Massi, M., Piomelli, D., Ciccocioppo,
ever, the ECS constitutes a clear therapeutic target for the treatment R., Rodriguez de Fonseca, F., 2007. The anandamide transport inhibitor AM404
reduces ethanol self-administration. Eur. J. Neurosci. 26, 476–486.
of alcohol dependence. In this way, the development of new specific Cippitelli, A., Bilbao, A., Hansson, A.C., del Arco, I., Sommer, W., Heilig, M., Massi,
cannabinoid antagonists could contribute to develop new pharma- M., Bermudez-Silva, F.J., Navarro, M., Ciccocioppo, R., Rodriguez de Fonseca, F.,
A.M. Erdozain, L.F. Callado / Drug and Alcohol Dependence 117 (2011) 102–110 109

2005. Cannabinoid CB1 receptor antagonism reduces conditioned reinstatement and substance dependence. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 141B,
of ethanol-seeking behavior in rats. Eur. J. Neurosci. 21, 2243–2251. 499–503.
Cohen, C., Perrault, G., Voltz, C., Steinberg, R., Soubrie, P., 2002. SR141716, a Houchi, H., Babovic, D., Pierrefiche, O., Ledent, C., Daoust, M., Naassila, M., 2005.
central cannabinoid (CB(1)) receptor antagonist, blocks the motivational and CB1 receptor knockout mice display reduced ethanol-induced conditioned place
dopamine-releasing effects of nicotine in rats. Behav. Pharmacol. 13, 451–463. preference and increased striatal dopamine D2 receptors. Neuropsychopharma-
Colombo, G., Agabio, R., Fa, M., Guano, L., Lobina, C., Loche, A., Reali, R., Gessa, G.L., cology 30, 339–349.
1998. Reduction of voluntary ethanol intake in ethanol-preferring sP rats by the Hungund, B.L., Basavarajappa, B.S., 2000. Distinct differences in the cannabinoid
cannabinoid antagonist SR-141716. Alcohol Alcohol. 33, 126–130. receptor binding in the brain of C57BL/6 and DBA/2 mice, selected for their
Colombo, G., Serra, S., Brunetti, G., Gomez, R., Melis, S., Vacca, G., Carai, M.M., Gessa, L., differences in voluntary ethanol consumption. J. Neurosci. Res. 60, 122–128.
2002. Stimulation of voluntary ethanol intake by cannabinoid receptor agonists Hungund, B.L., Szakall, I., Adam, A., Basavarajappa, B.S., Vadasz, C., 2003. Cannabinoid
in ethanol-preferring sP rats. Psychopharmacology (Berl.) 159, 181–187. CB1 receptor knockout mice exhibit markedly reduced voluntary alcohol con-
Colombo, G., Vacca, G., Serra, S., Carai, M.A., Gessa, G.L., 2004. Suppressing effect of sumption and lack alcohol-induced dopamine release in the nucleus accumbens.
the cannabinoid CB1 receptor antagonist SR 141716, on alcohol’s motivational J. Neurochem. 84, 698–704.
properties in alcohol-preferring rats. Eur. J. Pharmacol. 498, 119–123. Hutchison, K.E., Haughey, H., Niculescu, M., Schacht, J., Kaiser, A., Stitzel, J., Horton,
Comings, D.E., Muhleman, D., Gade, R., Johnson, P., Verde, R., Saucier, G., MacMurray, W.J., Filbey, F., 2008. The incentive salience of alcohol: translating the effects of
J., 1997. Cannabinoid receptor gene (CNR1): association with i.v. drug use. Mol. genetic variant in CNR1. Arch. Gen. Psychiatry 65, 841–850.
Psychiatry 2, 161–168. Ishiguro, H., Iwasaki, S., Teasenfitz, L., Higuchi, S., Horiuchi, Y., Saito, T., Arinami, T.,
De Bruin, N.M.W.J., Lange, J.H.M., Kruse, C.G., Herremans, A.H., Schoffelmeer, A.N.M., Onaivi, E.S., 2007. Involvement of cannabinoid CB2 receptor in alcohol prefer-
van Drimmelen, M., De Vries, T.J., 2011. SLV330 a cannabinoid CB1 receptor ence in mice and alcoholism in humans. Pharmacogenomics J. 7, 380–385.
antagonist, attenuates ethanol and nicotine seeking and improves inhibitory Iwasaki, S., Ishiguro, H., Higuchi, S., Onaivi, E.S., Arinami, T., 2007. Association study
response control in rats. Behav. Brain Res. 217, 408–415. between alcoholism and endocannabinoid metabolic enzyme genes encoding
Diamond, I., Gordon, A.S., 1997. Cellular and molecular neuroscience of alcoholism. fatty acid amide hydrolase and monoglyceride lipase in a Japanese population.
Physiol. Rev. 77, 1–20. Psychiatr. Genet. 17, 215–220.
Dyr, W., Ligieza, J., Kostowski, W., 2008. The effect of cannabinoid CB(1) receptor Kelai, S., Hanoun, N., Aufrere, G., Beauge, F., Hamon, M., Lanfumey, L., 2006.
antagonist rimonabant (SR-141716) on ethanol drinking in high-preferring rats. Cannabinoid–serotonin interactions in alcohol-preferring vs. alcohol-avoiding
Alcohol 42, 509–512. mice. J. Neurochem. 99, 308–320.
European Medicines Agency, 2009. Public Statement on Zimulti (Rimona- Köhnke, M.D., 2008. Approach to the genetics of alcoholism: a review based on
bant) Withdrawal of the Marketing Authorisation in the European Union pathophysiology. Biochem. Pharmacol. 75, 160–177.
(accessed on 2010/10/18). European Medicines Agency EMEA/39560/2009 Koob, G.F., Volkow, N.D., 2010. Neurocircuitry of addiction. Neuropsychopharma-
http://www.ema.europa.eu/humandocs/PDFs/EPAR/zimulti/3956009en.pdf. cology 35, 217–238.
Ferrer, B., Bermudez-Silva, F.J., Bilbao, A., Alvarez-Jaimes, L., Sanchez-Vera, I., Giuf- Lallemand, F., De Witte, P., 2005. Ethanol induces higher BEC in CB1 cannabinoid
frida, A., Serrano, A., Baixeras, E., Khaturia, S., Navarro, M., Parsons, L.H., Piomelli, receptor knockout mice while decreasing ethanol preference. Alcohol Alcohol.
D., Rodriguez de Fonseca, F., 2007. Regulation of brain anandamide by acute 40, 54–62.
administration of ethanol. Biochem. J. 404, 97–104. Lallemand, F., De Witte, P., 2006. SR147778, a CB1 cannabinoid receptor antagonist,
Flanagan, J.M., Gerber, A.L., Cadet, J.L., Beutler, E., Sipe, J.C., 2006. The fatty acid suppresses ethanol preference in chronically alcoholized Wistar rats. Alcohol
amide hydrolase 385 A/A (P129T) variant: haplotype analysis of an ancient 39, 125–134.
missense mutation and validation of risk for drug addiction. Hum. Genet. 120, Lallemand, F., Soubrie, P.H., De Witte, P.H., 2001. Effects of CB1 cannabinoid receptor
581–588. blockade on ethanol preference after chronic ethanol administration. Alcohol
Freedland, C.S., Sharpe, A.L., Samson, H.H., Porrino, L.J., 2001. Effects of SR141716A Clin. Exp. Res. 25, 1317–1323.
on ethanol and sucrose self-administration. Alcohol. Clin. Exp. Res. 25, 277–282. Lehtonen, M., Storvik, M., Tupala, E., Hyytia, P., Tiihonen, J., Callaway, J.C., 2010.
Galiegue, S., Mary, S., Marchand, J., Dussossoy, D., Carriere, D., Carayon, P., Bouaboula, Endogenous cannabinoids in post-mortem brains of Cloninger type 1 and 2
M., Shire, D., Le Fur, G., Casellas, P., 1995. Expression of central and peripheral alcoholics. Eur. Neuropsychopharmacol. 20, 245–252.
cannabinoid receptors in human immune tissues and leukocyte subpopulations. Lupica, C.R., Riegel, A.C., 2005. Endocannabinoid release from midbrain dopamine
Eur. J. Biochem. 232, 54–61. neurons: a potential substrate for cannabinoid receptor antagonist treatment of
Gallate, J.E., McGregor, I.S., 1999. The motivation for beer in rats: effects of ritanserin, addiction. Neuropharmacology 48, 1105–1116.
naloxone and SR 141716. Psychopharmacology (Berl.) 142, 302–308. Maldonado, R., Valverde, O., Berrendero, F., 2006. Involvement of the endocannabi-
Gallate, J.E., Saharov, T., Mallet, P.E., McGregor, I.S., 1999. Increased motivation for noid system in drug addiction. Trends Neurosci. 29, 225–232.
beer in rats following administration of a cannabinoid CB1 receptor agonist. Eur. Matsuda, L.A., Bonner, T.I., Lolait, S.J., 1993. Localization of cannabinoid receptor
J. Pharmacol. 370, 233–240. mRNA in rat brain. J. Comp. Neurol. 327, 535–550.
George, D.T., Herion, D.W., Jones, C.L., Phillips, M.J., Hersh, J., Hill, D., Heilig, M., Matsuda, L.A., Lolait, S.J., Brownstein, M.J., Young, A.C., Bonner, T.I., 1990. Structure
Ramchandani, V.A., Geyer, C., Spero, D.E., Singley, D.E., O’Malley, S.S., Bishai, R., of a cannabinoid receptor and functional expression of the cloned cDNA. Nature
Rawlings, R.R., Kunos, G., 2010. Rimonabant (SR141716) has no effect on alco- 346, 561–564.
hol self-administration or endocrine measures in nontreatment-seeking heavy Mitrirattanakul, S., Lopez-Valdes, H.E., Liang, J., Matsuka, Y., Mackie, K., Faull, K.F.,
alcohol drinkers. Psychopharmacology (Berl.) 208, 37–44. Spigelman, I., 2007. Bidirectional alterations of hippocampal cannabinoid 1
Gessa, G.L., Serra, S., Vacca, G., Carai, M.A., Colombo, G., 2005. Suppressing effect receptors and their endogenous ligands in a rat model of alcohol withdrawal
of the cannabinoid CB1 receptor antagonist SR147778, on alcohol intake and and dependence. Alcohol. Clin. Exp. Res. 31, 855–867.
motivational properties of alcohol in alcohol-preferring sP rats. Alcohol Alcohol. Moranta, D., Esteban, S., Garcia-Sevilla, J.A., 2006. Ethanol desensitizes cannabi-
40, 46–53. noid CB1 receptors modulating monoamine synthesis in the rat brain in vivo.
Glass, M., Dragunow, M., Faull, R.L., 1997. Cannabinoid receptors in the human Neurosci. Lett. 392, 58–61.
brain: a detailed anatomical and quantitative autoradiographic study in the fetal, Munro, S., Thomas, K.L., bu-Shaar, M., 1993. Molecular characterization of a periph-
neonatal and adult human brain. Neuroscience 77, 299–318. eral receptor for cannabinoids. Nature 365, 61–65.
Gonzalez, S., Cascio, M.G., Fernandez-Ruiz, J., Fezza, F., Di Marzo, V., Ramos, J.A., Naassila, M., Pierrefiche, O., Ledent, C., Daoust, M., 2004. Decreased alcohol self-
2002a. Changes in endocannabinoid contents in the brain of rats chronically administration and increased alcohol sensitivity and withdrawal in CB1 receptor
exposed to nicotine, ethanol or cocaine. Brain Res. 954, 73–81. knockout mice. Neuropharmacology 46, 243–253.
Gonzalez, S., Fernandez-Ruiz, J., Sparpaglione, V., Parolaro, D., Ramos, J.A., 2002b. Oliva, J.M., Ortiz, S., Perez-Rial, S., Manzanares, J., 2008. Time dependent alterations
Chronic exposure to morphine, cocaine or ethanol in rats produced different on tyrosine hydroxylase, opioid and cannabinoid CB1 receptor gene expressions
effects in brain cannabinoid CB(1) receptor binding and mRNA levels. Drug after acute ethanol administration in the rat brain. Eur. Neuropsychopharmacol.
Alcohol Depend. 66, 77–84. 18, 373–382.
Gonzalez, S., Valenti, M., de Miguel, R., Fezza, F., Fernandez-Ruiz, J., Di Marzo, V., Ortiz, S., Oliva, J.M., Perez-Rial, S., Palomo, T., Manzanares, J., 2004a. Chronic ethanol
Ramos, J.A., 2004. Changes in endocannabinoid contents in reward-related brain consumption regulates cannabinoid CB1 receptor gene expression in selected
regions of alcohol-exposed rats, and their possible relevance to alcohol relapse. regions of rat brain. Alcohol Alcohol. 39, 88–92.
Br. J. Pharmacol. 143, 455–464. Ortiz, S., Oliva, J.M., Perez-Rial, S., Palomo, T., Manzanares, J., 2004b. Differences in
Hansson, A.C., Bermudez-Silva, F.J., Malinen, H., Hyytia, P., Sanchez-Vera, I., Rimon- basal cannabinoid CB1 receptor function in selective brain areas and vulnerabil-
dini, R., Rodriguez de Fonseca, F., Kunos, G., Sommer, W.H., Heilig, M., 2007. ity to voluntary alcohol consumption in Fawn Hooded and Wistar rats. Alcohol
Genetic impairment of frontocortical endocannabinoid degradation and high Alcohol. 39, 297–302.
alcohol preference. Neuropsychopharmacology 32, 117–126. Ponce, G., Hoenicka, J., Rubio, G., Ampuero, I., Jimenez-Arriero, M.A., Rodriguez-
Hariri, A.R., Gorka, A., Hyde, L.W., Kimak, M., Halder, I., Ducci, F., Ferrell, R.E., Jimenez, R., Palomo, T., ramos, J.A., 2003. Association between cannabinoid
Goldman, D., Manuck, S.B., 2009. Divergent effects of genetic variation in endo- receptor gene (CNR1) and childhood attention deficit/hyperactivity disorder in
cannabinoid signaling on human threat- and reward-related brain function. Biol. Spanish male alcoholic patients. Mol. Psychiatry 8, 466–467.
Psychiatry 66, 9–16. Poncelet, M., Maruani, J., Calassi, R., Soubrie, P., 2003. Overeating, alcohol and
Herkenham, M., Lynn, A.B., Johnson, M.R., Melvin, L.S., de Costa, B.R., Rice, K.C., sucrose consumption decrease in CB1 receptor deleted mice. Neurosci. Lett. 343,
1991. Characterization and localization of cannabinoid receptors in rat brain: 216–218.
a quantitative in vitro autoradiographic study. J. Neurosci. 11, 563–583. Preuss, U.W., Koller, G., Zill, P., Bondy, B., Soyka, M., 2003. Alcoholism-related phe-
Herman, A.I., Kranzler, H.R., Cubells, J.F., Gelernter, J., Covault, J., 2006. Association notypes and genetic variants of the CB1 receptor. Eur. Arch. Psychiatry Clin.
study of the CNR1 gene exon 3 alternative promoter region polymorphisms Neurosci. 253, 275–280.
110 A.M. Erdozain, L.F. Callado / Drug and Alcohol Dependence 117 (2011) 102–110

Racz, I., Bilkei-Gorzo, A., Toth, Z.E., Michel, K., Palkovits, M., Zimmer, A., 2003. A Thanos, P.K., Dimitrakakis, E.S., Rice, O., Gifford, A., Volkow, N.D., 2005. Ethanol self-
critical role for the cannabinoid CB1 receptors in alcohol dependence and stress- administration and ethanol conditioned place preference are reduced in mice
stimulated ethanol drinking. J. Neurosci. 23, 2453–2458. lacking cannabinoid CB1 receptors. Behav. Brain Res. 164, 206–213.
Rinaldi-Carmona, M., Barth, F., Congy, C., Martinez, S., Oustric, D., Perio, A., Tsou, K., Nogueron, M.I., Muthian, S., Sanudo-Pena, M.C., Hillard, C.J., Deutsch, D.G.,
Poncelet, M., Maruani, J., Arnone, M., Finance, O., Soubrie, P., Le Fur, Walker, J.J., 1998. Fatty acid amide hydrolase is located preferentially in large
G., 2004. SR147778 [5-(4-bromophenyl)-1-(2,4-dichlorophenyl)-4-ethyl-N- neurons in the rat central nervous system as revealed by immunohistochem-
(1-piperidinyl)-1H-pyr azole-3-carboxamide], a new potent and selective istry. Neurosci. Lett. 254, 137–140.
antagonist of the CB1 cannabinoid receptor: biochemical and pharmacological Vinod, K.Y., Arango, V., Xie, S., Kassir, S.A., Mann, J.J., Cooper, T.B., Hungund, B.L., 2005.
characterization. J. Pharmacol. Exp. Ther. 310, 905–914. Elevated levels of endocannabinoids and CB1 receptor-mediated G-protein sig-
Rodriguez de Fonseca, F., Roberts, A.J., Bilbao, A., Koob, G.F., Navarro, M., 1999. naling in the prefrontal cortex of alcoholic suicide victims. Biol. Psychiatry 57,
Cannabinoid receptor antagonist SR141716A decreases operant ethanol self 480–486.
administration in rats exposed to ethanol-vapor chambers. Zhongguo Yao Li Vinod, K.Y., Kassir, S.A., Hungund, B.L., Cooper, T.B., Mann, J.J., Arango, V., 2010. Selec-
Xue Bao 20, 1109–1114. tive alterations of the CB1 receptors and the fatty acid amide hydrolase in the
Rubio, M., de Miguel, R., Fernandez-Ruiz, J., Gutierrez-Lopez, D., Carai, M.A., ventral striatum of alcoholics and suicides. J. Psychiatr. Res. 44, 591–597.
Ramos, J.A., 2009. Effects of a short-term exposure to alcohol in rats on FAAH Vinod, K.Y., Sanguino, E., Yalamanchili, R., Manzanares, J., Hungund, B.L., 2008.
enzyme and CB1 receptor in different brain areas. Drug Alcohol Depend. 99, Manipulation of fatty acid amide hydrolase functional activity alters sensitivity
354–358. and dependence to ethanol. J. Neurochem. 104, 233–243.
Rubio, M., McHugh, D., Fernandez-Ruiz, J., Bradshaw, H., Walker, J.M., 2007. Short- Vinod, K.Y., Yalamanchili, R., Xie, S., Cooper, T.B., Hungund, B.L., 2006. Effect of
term exposure to alcohol in rats affects brain levels of anandamide, other N- chronic ethanol exposure and its withdrawal on the endocannabinoid system.
acylethanolamines and 2-arachidonoyl-glycerol. Neurosci. Lett. 421, 270–274. Neurochem. Int. 49, 619–625.
Schmidt, L.G., Samochowiec, J., Finckh, U., Fiszer-Piosik, E., Horodnicki, J., Wendel, B., Wang, G.J., Volkow, N.D., Franceschi, D., Fowler, J.S., Thanos, P.K., Scherbaum, N., Pap-
Rommelspacher, H., Hoehe, M.R., 2002. Association of a CB1 cannabinoid recep- pas, N., Wong, C.T., Hitzemann, R.J., Felder, C.A., 2000. Regional brain metabolism
tor gene (CNR1) polymorphism with severe alcohol dependence. Drug Alcohol during alcohol intoxication. Alcohol. Clin. Exp. Res. 24, 822–829.
Depend. 65, 221–224. Wang, L., Liu, J., Harvey-White, J., Zimmer, A., Kunos, G., 2003. Endocannabinoid
Serra, S., Brunetti, G., Pani, M., Vacca, G., Carai, M.A., Gessa, G.L., Colombo, G., 2002. signaling via cannabinoid receptor 1 is involved in ethanol preference and its
Blockade by the cannabinoid CB(1) receptor antagonist SR 141716, of alcohol age-dependent decline in mice. Proc. Natl. Acad. Sci. U.S.A. 100, 1393–1398.
deprivation effect in alcohol-preferring rats. Eur. J. Pharmacol. 443, 95–97. World Health Organization, 2004. Global Status Report on Alcohol 2004. World
Serra, S., Carai, M.A., Brunetti, G., Gomez, R., Melis, S., Vacca, G., Colombo, G., Gessa, Health Organization, Geneva.
G.L., 2001. The cannabinoid receptor antagonist SR 141716 prevents acquisition Wilson, R.I., Nicoll, R.A., 2002. Endocannabinoid signaling in the brain. Science 296,
of drinking behavior in alcohol-preferring rats. Eur. J. Pharmacol. 430, 369–371. 678–682.
Sipe, J.C., Chiang, K., Gerber, A.L., Beutler, E., Cravatt, B.F., 2002. A missense mutation Zhang, P.W., Ishiguro, H., Ohtsuki, T., Hess, J., Carillo, F., Walther, D., Onaivi, E.S.,
in human fatty acid amide hydrolase associated with problem drug use. Proc. Arinami, T., Uhl, G.R., 2004. Human cannabinoid receptor 1: 5 exons, candidate
Natl. Acad. Sci. U.S.A. 99, 8394–8399. regulatory regions, polymorphisms, haplotypes and association with polysub-
Soyka, M., Koller, G., Schmidt, P., Lesch, O.M., Leweke, M., Fehr, C., Gann, H., Mann, stance abuse. Mol. Psychiatry 9, 916–931.
K.F., 2008. Cannabinoid receptor 1 blocker rimonabant (SR 141716) for treat- Zuo, L., Kranzler, H.R., Luo, X., Covault, J., Gelernter, J., 2007. CNR1 variation modu-
ment of alcohol dependence: results from a placebo-controlled, double-blind lates risk for drug and alcohol dependence. Biol. Psychiatry 62, 616–626.
trial. J. Clin. Psychopharmacol. 28, 317–324.

You might also like