You are on page 1of 60

Behavioural Pharmacology

The endocannabinoid system and drug-associated contextual memories


--Manuscript Draft--

Manuscript Number: BP-20-91

Full Title: The endocannabinoid system and drug-associated contextual memories

Article Type: Review Article

Section/Category: Non-United States/Canada

Keywords: Cannabinoids; reward; addiction; drug abuse; memory; conditioned place


preference

Corresponding Author: Fabricio A. Moreira, PhD


University of Sao Paulo
Ribeirao Preto, BRAZIL

Corresponding Author Secondary


Information:

Corresponding Author's Institution: University of Sao Paulo

Corresponding Author's Secondary


Institution:

First Author: Laila Asth

First Author Secondary Information:

Order of Authors: Laila Asth

Aline C. Santos

Fabricio A. Moreira, PhD

Order of Authors Secondary Information:

Manuscript Region of Origin: BRAZIL

Abstract: Drug abuse and addiction can be initiated and reinstated by contextual stimuli
previously paired with the drug use. The influence exerted by the context on drug
seeking behaviour can be modelled in experimental animals with place conditioning
protocols. Here we review the effects of cannabinoids in place conditioning and the
therapeutic potential of the endocannabinoid system for interfering with drug-related
memories. The phytocannabinoid delta 9 -tetrahydrocannabinol (THC) tends induce
CPP at low doses and CPA at high doses; cannabidiol is devoid of any effect and yet it
can inhibit CPP induced by some drugs. Synthetic CB 1 receptor agonists tend to
recapitulate the biphasic profile observed with THC, whereas selective
antagonists/inverse agonists inhibit CPP induced by cocaine, nicotine, alcohol and
opioids. The CB 2 receptor has also attracted attention, as selective CB 2 agonists
inhibit cocaine-induced CPP. Inhibitors of endocannabinoid membrane transport and
hydrolysis yield mixed results. In targeting the endocannabinoid system for developing
new treatments for drug addiction, future research should focus on “neutral” CB 1
receptor antagonists and CB 2 receptor agonists. Such compounds may offer a safe
pharmacological profile and curb addiction by preventing drug seeking triggered by
conditioned contextual cues.

Powered by Editorial Manager® and ProduXion Manager® from Aries Systems Corporation
Covering Letter

___________________________________________________________________________
Universidade Federal de Minas Gerais (UFMG)
Instituto de Ciências Biológicas | Departamento de Farmacologia

Belo Horizonte, Brazil


06th May 2020

Dear Dr. Louk Vanderschuren, Editor of Behavioural Pharmacology

We are submitting the manuscript

The endocannabinoid system and drug-associated contextual memories

To be considered for publication in the special issue Translational Research in Behavioural


Pharmacology.

All authors have contributed to this work and have agreed on the final version of the
manuscript. Also, this manuscript has not been submitted to any other journal. We have no
conflict of interest to declare.

Thank you and best regards,

Fabricio A. Moreira, PhD


Department of Pharmacology, ICB, Federal University of Minas Gerais
Av. Antonio Carlos 6627; 31270-901 Belo Horizonte, MG; Brazil
Tel.: +55 31 3409 2720; E-mail: fabriciomoreira@icb.ufmg.br
Manuscript (All Manuscript Text Pages, including Title Page,
References and Figure Legends)

Review paper

The endocannabinoid system and drug-associated contextual memories

Running head: Cannabinoids and drug memories

Laila ASTH#, Aline C. SANTOS#, Fabrício A. MOREIRA*

Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de

Minas Gerais, Belo Horizonte, MG, Brazil, 31270-901.

#
Shared first authorship

*Corresponding author:

Dr. Fabrício de Araújo Moreira, Department of Pharmacology, Institute of Biological

Sciences, Universidade Federal de Minas Gerais. Av. Antônio Carlos 6627, Belo

Horizonte, MG, 31270-901, Brazil. E-mail: fabriciomoreira@icb.ufmg.br

Conflicts of interest

None.

1
Acknowledgements

LA thanks “Coordenação de Aperfeiçoamento de Pessoal de Nível Superior – Brasil

(CAPES) – Finance Code 001” for the post-doctoral fellowship. ACS thanks “The

Brazilian National Council for Scientific and Technological Development (CNPq)” for

a master degree fellowship. FAM thanks “The Brazilian National Council for Scientific

and Technological Development (CNPq)” for the research grant 406122/2016-4 and for

the research productivity fellowship (level 2); “Fundação de Amparo à Pesquisa do

Estado de Minas Gerais” (FAPEMIG), for the research grant APQ-02064-15;

“Fundação de Amparo à Pesquisa do Estado de São Paulo” (FAPESP, Projeto Temático

2017/24304-0) and Instituto Nacional de Ciência e Tecnologia (INCT-Translational

Medicine).

2
Abstract

Drug abuse and addiction can be initiated and reinstated by contextual stimuli

previously paired with the drug use. The influence exerted by the context on drug

seeking behaviour can be modelled in experimental animals with place conditioning

protocols. Here we review the effects of cannabinoids in place conditioning and the

therapeutic potential of the endocannabinoid system for interfering with drug-related

memories. The phytocannabinoid 9-tetrahydrocannabinol (THC) tends induce CPP at

low doses and CPA at high doses; cannabidiol is devoid of any effect and yet it can

inhibit CPP induced by some drugs. Synthetic CB1 receptor agonists tend to recapitulate

the biphasic profile observed with THC, whereas selective antagonists/inverse agonists

inhibit CPP induced by cocaine, nicotine, alcohol and opioids. The CB2 receptor has

also attracted attention, as selective CB2 agonists inhibit cocaine-induced CPP.

Inhibitors of endocannabinoid membrane transport and hydrolysis yield mixed results.

In targeting the endocannabinoid system for developing new treatments for drug

addiction, future research should focus on “neutral” CB1 receptor antagonists and CB2

receptor agonists. Such compounds may offer a safe pharmacological profile and curb

addiction by preventing drug seeking triggered by conditioned contextual cues.

Key words: Cannabinoids; reward; addiction; drug abuse; memory; conditioned place

preference

3
INTRODUCTION

Several substances consumed by humans, including ethanol (alcohol), opioids, cocaine,

amphetamines, nicotine and caffeine, serve as rewarding and reinforcing stimuli leading

some individuals to compulsive abuse and addiction (Everitt and Robbins, 2005). Drug

addiction is considered as the most severe form of substance use disorder, which entails

several components, including craving, use of a substance despite all related problems

and negative consequences for everyday life, pharmacological tolerance, withdrawal

symptoms and relapse (DSM-5). Substance use disorder represents as a world health

problem whose prevalence has been increasing over the years (Collaborators, 2018).

Despite its huge impact on individual health and society, few effective treatments exist

(O'Brien, 2008,Welberg, 2011). A major challenge in developing addiction

pharmacotherapies can be attributed to the continuous change in brain circuitry after

chronic exposure to drugs of abuse. One conceptual framework divides drug addiction

in three phases: binge and intoxication, withdrawal and negative affect, and

preoccupation and anticipation (or craving) leading to relapse, which perpetuates drug

use (Koob and Le Moal, 2008,Koob and Volkow, 2016). Acute administration of drugs

of abuse facilitates dopamine neurotransmission in the reward mesolimbic pathway,

which projects from ventral tegmental area (VTA) to the nucleus accumbens (NAcc) in

the ventral striatum (Di Chiara and Imperato, 1988). As drug use progress toward

addiction, molecular and cellular modifications occur in the mesolimbic system, dorsal

aspects of the basal ganglia, extended amygdala, hippocampus and subregions of the

prefrontal cortex (Everitt and Robbins, 2005,Wise and Koob, 2014,Koob and Volkow,

2016).

4
The memory related to the context in which drug is consumed also constitutes a key

feature in the process from initial drug use towards addiction. A previously neutral

context and its related cues may become rewarding (conditioned stimuli) once they are

associated with drugs of abuse (interoceptive unconditioned stimuli), precipitating drug-

seeking behaviour and relapse. Therefore, drug addiction has been proposed as a

disorder whose investigation should take into account the subjacent memory systems

(Milton and Everitt, 2012). Specific sub-nucleus of the amygdala seem to be necessary

for processing emotional memories associated with drug use; the hippocampus, in turn,

is required for the acquisition and retrieval of contextual information previously paired

with the drug, which in turn can perpetuate drug seeking through neuronal connections

with the mesolimbic reward pathway (Milton and Everitt, 2012).

Several neurotransmitter systems are thought to be involved in addiction and substance

use disorders. Among them, the endocannabinoid system stands as a particularly

interesting neurochemical mechanism, controlling brain mechanisms underlying both

contextual memory and reward. This review focuses primarily on the cannabinoid link

between contextual memory and rewarding properties of drugs. First, we will discuss

the place conditioning protocol, emphasizing its translational value, strengths and

limitations. Next, we provide a brief overview on the role of the endocannabinoid

system in reward and addiction. The main session will focus on the relevance of place

conditioning to investigate the effects of cannabinoids and compounds targeting the

endocannabinoid system as potential interventions for drug-related contextual

memories. Finally, we will provide a summary of the main picture emerging from these

studies and suggest directions for further pharmacological research.

5
CONDITIONED PLACE PREFERENCE

Considering the need for better understanding the neurobiology drug addiction and

developing new pharmacotherapies, experimental studies with laboratory animals have

been extensively used (Kuhn et al., 2019). Of special interest for investigating drug-

related contextual memories is the conditioned place preference protocol (CPP), which

was first validated for studying opioids (Katz and Gormezano, 1979) and have been

extensively used for several drugs (Bardo and Bevins, 2000,Cunningham et al.,

2006,Huston et al., 2013). CPP is a simple and reliable procedure, which can be used

with mice and rats as experimental animals. A schematic overview of the apparatus and

the protocol is depicted in Figure 1. The apparatus consists of two chambers with

different visual and tactile cues, which can be connected by a small third compartment.

Although protocols may vary across studies, a typical CPP study consists in at least

three phases. First, animals are exposed to a pre-conditioning session, in which they are

allowed to explore the apparatus (two or three compartments). Then, animals undergo

the conditioned phase. The drug (unconditioned stimulus) is administered and the

animal is immediately confined to one of the chambers (which will serve as the

contextual conditioned stimulus). In alternate sessions, the vehicle is administered and

paired with the opposite chamber. During this phase, animals learn to associate the

environment with reward (or aversive) interoceptive effects induced by the drug.

Finally, in the test, drug-free animals are again able to explore the entire apparatus. If

they spend more time in the compartment previously paired to the drug than in unpaired

one, the drugs is considered as rewarding (CPP), while if the opposite occurs, the drug

effect is interpreted as aversive (conditioned place aversion, CPA) (Bardo and Bevins,

2000,Cunningham et al., 2006,Tzschentke, 2007,Huston et al., 2013). Place

conditioning (CPP or CPA) probably reflects the animals seeking (or avoiding) the drug

6
in the environment associated with its rewarding (or aversive) effects. In extended

protocols, reconsolidation, extinction and reinstatement can also be investigated (Parker

and Mcdonald, 2000). Reconsolidation can be evaluated when animals undergo a drug

memory reactivation phase followed by a retesting of drug-induced conditioned place

preference or aversion. Extinction is achieved by exposuring animals to paired

compartment without drug injection and reinstatement is induced by drug-priming

injections or exposure to stressors (Liu et al., 2008). In this way, it is also possible to

evaluate relapse to drug-seeking behaviour using CPP.

CPP has some important limitations (Bardo and Bevins, 2000). A pharmacological

limitation concerns the difficult in generating graded dose-effect curves due to both the

nature of the response and the requirement for a between-groups experimental design.

Another major problem is the broad variation in protocols used in different laboratories.

Some studies report no initial preference by the animal for any compartment of the

apparatus in the pre-conditioning session (unbiased protocol), while others report

preference for one of the sides (biased protocol). In case of a biased protocol, the reward

stimulus can be paired preferentially with the non-preferred compartment. Other source

of cross-studies variation are the duration and number of exposures to the apparatus.

However, the most obvious drawback of CPP concerns the drug administration by the

experimenter, rather than by the animal itself. In this way, the protocol fails to

recapitulate the contingency between the operant response (the drug self-administration)

and the positive reinforcing properties of the drug (leading to subsequent drug seeking),

an important feature of addiction.

In spite of these limitations, several advantages and applications are ascribed to place

conditioning protocols. The experiments can be easily performed with high

7
reproducibility, allowing investigation of both preference (CPP) and aversion (CPA).

Moreover, CPP exhibits robust face, construct and predictive validities (Bardo and

Bevins, 2000). CPP face validity relies on the possibility of analysing drug seeking

behaviour triggered by a conditioned reward, the context. Although this seems less

straightforward as compared to the face validity of self-administration protocols, CPP

can be induced also in humans by contextual cues associated with drugs (Childs and de

Wit, 2009,2013) as well as music (Molet et al., 2013), food (Astur et al., 2014), and

money (Childs et al., 2017). Furthermore, the exposure of animals to different types of

stressors induces reinstatement of CPP (Mantsch et al., 2016), modelling observations

in humans in which aversive events motivate relapse to drugs of abuse (Sinha et al.,

2006,Back et al., 2010,Higley et al., 2011,Sinha et al., 2011). The construct validity of

this protocol is supported by studies demonstrating the increase in dopamine released in

forebrain areas after the exposition to a drug-related cue in both animals and humans

(Milella et al., 2016,Shinohara et al., 2017). Moreover, brain structures and circuits

responsible for processing contextual information paired with drugs of abuse, such as

the hippocampus and its connections to the NAcc, have been demonstrated to underly

CPP (LeGates et al., 2018,Sjulson et al., 2018).

As for its pharmacological predictability, various drugs have been investigated in mice

and rats in CPP protocols. Among the drugs of abuse consistently reported to induce

conditioned reward are opioids, alcohol, amphetamines, cocaine, caffeine and nicotine

(Tzschentke, 1998,Liu et al., 2008). The protocol has also been demonstrated in humans

for amphetamine (Childs and de Wit, 2009,2013) and alcohol (Childs and de Wit,

2016). Apart from drugs, CPP can also be induced by food, sex and social interaction as

unconditioned stimuli (Agmo et al., 1995,Cunningham et al., 2006). Thus, CPP has a

robust predictive validity for detecting stimuli which are rewarding in humans. In

8
addition, CPP has been extensively used in the search for new pharmacological

treatments for addiction. The testing compound can be investigated for its effect upon

conditioning (acquisition), by administrations before each injection of the drug of abuse,

expression, by administering the compound before the test, as well as reconsolidation,

extinction and reinstatement (Tzschentke, 2007).

THE ENDOCANNABINOID SYSTEM AND DRUG ADDICTION

Among the compounds investigated in CPP/CPA protocols are the cannabinoids and

those targeting various molecular components of the endocannabinoid system. This

neuromodulatory system is named after the herb Cannabis sativa, whose main active

constituent is Δ9-tetrahydrocannabinol (THC), in addition to cannabidiol (CBD) and

other compounds called phytocannabinoids. The structure and functioning of this

system have been extensively reviewed (Pertwee et al., 2010,Mechoulam and Parker,

2013,Mechoulam et al., 2014). Endocannabinoid signalling comprises at least two

metabotropic, Gi protein-compled receptors, type 1 (CB1) and type 2 (CB2) cannabinoid

receptors (Matsuda et al., 1990,Munro et al., 1993); their endogenous ligands

(endocannabinoids), the lipids anandamide and 2-arachidonoylglycerol (2-AG) (Devane

et al., 1992,Sugiura et al., 1995); and their metabolizing enzymes. Endocannabinoids

are synthesized on-demand from postsynaptic membrane lipids after calcium influx and

subsequent activation of different enzymes, among them phospholipase D, for

anandamide, and diacylglycerol lipase α, for 2-AG (Bisogno et al., 2003,Okamoto et al.,

2004). As for their hydrolysis, anandamide and 2-AG hydrolysis are internalized in

neurons through a membrane transporter and serve as substrates primarily for fatty acid

amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), respectively (Cravatt

9
et al., 1996,Dinh et al., 2002). A schematic view of the endocannabinoid system is

presented in Figure 2.

The CB1 receptor is the main responsible for the typical effects of THC (a

phytocannabinoid) and its derivatives (synthetic cannabinoids). It is densely expressed

throughout the encephalon, including regions related to the development of various

aspects of reward and addiction, such as ventral and dorsal striatum, hippocampus,

amygdala and the prefrontal cortex (Herkenham et al., 1991,Tsou et al., 1998). Based

morphological, neurochemical and electrophysiological studies, the CB1 receptor is

proposed be located mainly in presynaptic terminals, with endocannabinoids being

synthesized from cell membranes in postsynaptic neurons. Therefore the

endocannabinoid system is considered as a retrograde neuromodulatory system, which

inhibit neuronal activity, firing rates and neurotransmitter release (Elphick and

Egertová, 2001,Kreitzer and Regehr, 2001,Ohno-Shosaku et al., 2001,Wilson and

Nicoll, 2001). Contrary to the CB1 receptor, CB2 expression and distribution in brain

has been controversial. It was initially proposed as a “peripheral” cannabinoid receptor,

absent from neurons in the encephalon, where it would be expressed only in glial cell

(Munro et al., 1993). More recent studies, however, have been challenging this view,

providing evidence for the expression of the CB2 receptor in neurons, including in

brains circuits related to addiction (Gong et al., 2006,Onaivi et al., 2008b,Zhang et al.,

2014,Zhang et al., 2017).

The connection between cannabinoids and addiction emerges from the well-known

abuse potential of cannabis preparations. Cannabis effects are mediated mainly by THC,

a phytocannabinoid with moderate efficacy as a CB1 and CB2 receptor agonist. Various

synthetic cannabinoids mimic THC effects, some of which possessing higher

10
selectivity, affinity, efficacy for the CB1 receptor than THC itself (Pertwee et al., 2010).

In experimental animals, cannabinoids tend to induce anxiolytic-like effects, sustain

self-administration, serve as unconditioned stimuli for CPP and facilitate mesolimbic

dopaminergic activity through CB1 receptor activation, at least when administered at

low doses. As a corollary, CB1 receptor antagonists consistently reduce rewarding and

reinforcing effects induced by cannabinoid and non-cannabinoid drugs, including

cocaine, ethanol, opioids and nicotine, suggesting a modulatory role for the

endocannabinoid system in drug addiction (Moreira et al., 2015,Parsons and Hurd,

2015,Manzanares et al., 2018). Accordingly, certain drugs of abuse, namely cocaine,

nicotine and ethanol, require CB1 receptor activation to induce dopamine release (Cheer

et al., 2007). Moreover, endocannabinoid-synthesizing enzymes are expressed in

dopaminergic neurons (Mátyás et al., 2008) and cocaine triggers the synthesis of 2-AG

in the VTA, which in turn activates CB1 to inhibits gamma-aminobutyric acid (GABA)

afferences and disinhibit dopaminergic neurons (Wang et al., 2015). In addition to the

VTA, 2-AG-mediated activation of CB1 receptor may also indirectly modulate

dopaminergic terminals the striatum (Uchigashima et al., 2007). Finally, the CB2

receptor has also been implicated in drug addiction, although its function seems to be

opposite to those ascribed to the CB1 receptor, at least for cocaine. CB2 receptor

agonists inhibit the behavioural effects of cocaine in mice after systemic or intra-NAcc

administrations (Xi et al., 2011,Zhang et al., 2014,Delis et al., 2017,Gobira et al.,

2019,Lopes et al., 2020). The mechanisms might involve direct modulation of

dopaminergic activity, since the CB2 receptor is co-expressed with tyrosine hydroxylase

in the VTA, inhibiting the activity dopamine-producing neurons (Zhang et al., 2017)

The expression and functioning of the endocannabinoid system point to an important

role of this neuro-modulatory system in pathways and brain regions underlying

11
contextual memory formation. Thus, place conditioning protocols have been extensively

used to investigate the reward or aversive characteristics of cannabinoids as well as the

role of the endocannabinoid system in the drug addiction.

CANNABINOIDS AND PLACE CONDITIONING

This section will report on the effects of cannabinoids and compounds targeting the

endocannabinoid system on acquisition (conditioning), expression (test), extinction,

reinstatement, and reconsolidation of reward memory. Table 1 shows the main ligands

employed in these studies along with their molecular targets. Their detailed

pharmacology has been reviewed elsewhere (Pertwee et al., 2010).

THC and CBD

Notwithstanding the well-known rewarding properties of cannabis, it has been difficult

to recapitulate these effects with systemic THC administration on place conditioning

protocols. Rewarding effects (CPP) tend to occur at low doses (Braida et al., 2004,Le

Foll et al., 2006) and depend not only on the CB1 receptor activation, but also on

recruitment of endogenous opioid mechanisms (Braida et al., 2004). In contrast,

aversion (CPA) is more commonly observed by increasing THC doses (Sañudo-Peña et

al., 1997,Hutcheson et al., 1998,Cheer et al., 2000,Quinn et al., 2008,DeVuono et al.,

2017). Various parameter may interfere with THC effects, since the aversiveness

induced by this drug is not observed in adolescent rats (Quinn et al., 2008) or when a

stress stimulus precedes the conditioning phase (DeVuono et al., 2017). Moreover, THC

12
effects on CPP/CPA are dependent on the previous experience of the subject with the

drug, since CPP has been attained, even at doses otherwise aversive to naïve subjects,

when the animals receive a previous THC administration (Valjent and Maldonado,

2000). Other studies reported no effects regardless the doses tested (Parker et al.,

2004,Vlachou et al., 2007,Hempel et al., 2016). Thus, studies with systemic

administration of THC report CPP most commonly at low doses (1 mg/kg, in average)

and CPA at high doses (around 5 mg/kg) although, in fact, the most frequent result is no

effect at all (Kubilius et al., 2018). Apart from systemic injections, THC induces CPP

after local administration into either the VTA or the NAcc (Zangen et al., 2006).

Regarding the interactions between THC and other drugs, this phytocannabinoid

facilitates the extinction of cocaine- and amphetamine-induced CPP (Parker et al.,

2004). In nicotine pre-exposed rats, low doses of THC does not induce CPP, whereas

higher doses induce CPA (Le Foll et al., 2006). In addition, protracted treatment with

THC followed by short and long abstinence periods prevent morphine-induced CPP

(Valverde et al., 2001,Jardinaud et al., 2006). THC also influences the response to other

cannabinoids. Previous THC administration5 reduced CPA induced by the potent

cannabinoid JWH-018, suggesting protection against the aversive effects of synthetic

cannabinoids (Hyatt and Fantegrossi, 2014).

CBD, a major cannabis constituent, binds to multiple targets in brain and exert a myriad

of pharmacological effects, among them anxiolytic, antipsychotic and antiepileptic (Izzo

et al., 2009). It seems to lack rewarding properties and fails to induce CPP or CPA per

se (Parker et al., 2004,Vann et al., 2008,Markos et al., 2018). However, it attenuates

acquisition of CPP induced by cocaine (Luján et al., 2018) and morphine (Markos et al.,

2018), and facilitates the extinction of cocaine- and amphetamine-induced CPP (Parker

et al., 2004). Moreover, CBD prevents drug- and stress-induced reinstatement of CPP to

13
morphine and cocaine (de Carvalho and Takahashi, 2017), as well as reinstatement of

CPP precipitated by methamphetamine (DeVuono et al., 2017). Yet, CBD inhibits CPA

precipitated by naltrexone (de Carvalho and Takahashi, 2017). Thus, CBD prevents

different phases of drug-induced CPP or CPA. CBD and THC have also been evaluated

in combination. CBD was able to prevent THC-induced CPA (Vann et al., 2008), an

observation with potential therapeutic implication since some cannabis-based medicines

contain a combination of both compounds. Therefore, these results highlight the

therapeutic potential of CBD for the treatment of drug addiction. In agreement, a pilot

study performed in tobacco smokers found reduction in the number of cigarettes

smoked after inhalation of CBD for one-week (Morgan et al., 2013) and a study with

opioid users observed that CBD reduces drug craving induced by conditioned cues

(Hurd et al., 2019).

CB1 receptor

The CB1 receptor used to be considered as the only cannabinoid receptor expressed in

neurons in the encephalon, accounting for the typical behavioural effects of THC and

synthetic cannabinoids. Both agonists and antagonists have been evaluated for its

effects on CPP/CPA. They have also been investigated on CPP induced by drugs of

abuse. In the same fashion as THC, synthetic cannabinoids (with different degrees of

CB1 selectivity) yield complex effects. Some authors observed CPP after systemic CB1

receptor activation (Braida et al., 2001), others reported CPA (Chaperon et al.,

1998,López-Moreno et al., 2004,Scherma et al., 2008a) and others found no effect

(Polissidis et al., 2009). However, intra-cranial injections into the NAcc (Karimi et al.,

2013) or VTA (Rashidy-Pour et al., 2013,Ahmad and Laviolette, 2017) induced CPP.

14
Overall, experiments with synthetic cannabinoids yield a profile similar to THC, with

CPP observed mainly at low doses and CPA at high doses (Kubilius et al., 2018).

CB1 receptor agonists have also been investigated in relation to their effects on CPP

induced by other drugs. They facilitate stress-induced reinstatement of cocaine CPP

when combined with an α-2 adrenergic antagonist, suggesting involvement of

noradrenergic pathways (Vaughn et al., 2012). Indeed, noradrenaline is one of the

neurotransmitters involved in negative emotional states associated with addiction (Koob

and Le Moal, 2008). CB1 receptor agonists also faciliate MDMA-induced reinstatement

of CPP by potentiating the effects of subthreshold priming doses of the drug (Daza-

Losada et al., 2011). Moreover, cannabinoid exposure during adolescence faciliate

reinstatement of MDMA-induced CPP in adult animals (Rodríguez-Arias et al., 2010).

These animals were also more susceptible to acquisition and reinstatement of CPP

induced by cocaine (Rodríguez-Arias et al., 2016). These results build on the hypothesis

that previous exposure to cannabinoids may lead to the subsequent use of other drugs

(Lynskey et al., 2003).

In contrast to agonists, CB1 receptor antagonists/inverse agonists generally fail to

induce any place preference or aversion per se (Mas-Nieto et al., 2001,Manzanedo et

al., 2004,Scherma et al., 2008a,Fang et al., 2011), although some studies do detect CPP

(Cheer et al., 2000,Ramiro-Fuentes et al., 2010) or CPA (Karimi et al., 2013).

Although CB1 receptor antagonists generally are devoid of effects on CPP/CPA

protocols, they consistently impair CPP induced by different classes of drugs,

particularly psychostimulants, which inhibit the uptake and/or facilitate the release of

dopamine. On cocaine-induced CPP, CB1 antagonism prevented acquisition (Chaperon

et al., 1998,Yu et al., 2011,Delis et al., 2017,Gobira et al., 2019,Lopes et al., 2020),

15
expression (Delis et al., 2017,Lopes et al., 2020), and reinstatement of reward memory

(Yu et al., 2011,Vaughn et al., 2012). We observed that the inhibitory effect of CB1

antagonists on cocaine CPP are reversed by a CB2 antagonist, suggesting an interplay

between cannabinoid receptors in modulating cocaine reward memories (Gobira et al.,

2019). CB1 antagonists also decrease CPP elicited by methamphetamine (Yu et al.,

2011), although they facilitate MDMA reinstatement of extinguished CPP (Daza-

Losada et al., 2011).

In addition to psychostimulants, CB1 antagonists have been investigated in CPP induced

by drugs acting through other mechanisms. They decreased acquisition (Le Foll and

Goldberg, 2004,Forget et al., 2005,Merritt et al., 2008), expression (Le Foll and

Goldberg, 2004), reinstatement (Budzyńska et al., 2009,Fang et al., 2011) and

reconsolidation (Fang et al., 2011) of nicotine-induced CPP. These consistent result are

supported by clinical trials showing the ameliorating effects of the CB1 receptor

antagonist rimonabant in nicotine dependence (Le Foll et al., 2008). They also impaired

acquisition (Chaperon et al., 1998,Mas-Nieto et al., 2001,Manzanedo et al., 2004,Singh

et al., 2004) and reconsolidation of CPP induced by morphine (De Carvalho et al.,

2014). These effects were mimicked with specific administration into the VTA or the

NAcc (Karimi et al., 2013,Rashidy-Pour et al., 2013). CB1 blockage is also effective in

CPP induced by other types of reward, particularly palatable food (Chaperon et al.,

1998,Méndez-Díaz et al., 2012).

The role of the CB1 receptor has also been investigated in genetically modified animals

(i.e. with receptor deletion or overexpression). The majority of the results converge with

data obtained using pharmacological approaches. CB1 knockout animals display

decreased CPP for ethanol (Houchi et al., 2005,Thanos et al., 2005), morphine (Martin

16
et al., 2000), and nicotine (Castañé et al., 2002,Merritt et al., 2008), although not for

cocaine and quinpirole (Martin et al., 2000,Castañé et al., 2002,Houchi et al., 2005). In

contrast, drug reward is not significantly changed in mice with a hyper-sensitive form of

CB1, since they develop CPP for ethanol, morphine, and cocaine (Marcus et al., 2017).

These discrepant results may be related to developmental changes in knock-out animals

in reward pathways (Houchi et al., 2005), which are modulated by CB1 receptor (Cheer

et al., 2007).

Therefore, CB1 receptor agonists induce either reward or aversive effects and facilitate

or inhibit CPP induced by other drugs, depending on the dose, protocol and

experimental settings. In contrast, CB1 receptor antagonists/inverse agonists are inert

per se, yet they consistently prevent all phases of CPP induced not only by drugs of

abuse, but also by other types of reward. The results from CB1 knockout mice tend to

confirm the conclusion from studies employing selective antagonist.

CB2 receptor

Contrary to the CB1 receptor, which accounts for most of the typical effects of

cannabinoids, data on CB2 expression in neurons and its possible role in behaviour are

still scant. CB2 receptor ligands are mostly devoid of reward or aversive properties on

place conditioning protocols. As for their effects on drug-induced CPP, contrasting with

CB1, it is the agonism (not the antagonism) of CB2 that prevents acquisition and

expression of cocaine-induced CPP (Delis et al., 2017,Lopes et al., 2020). Either CB1

antagonism or CB2 agonism prevented the increase in cFos expression in the

hippocampus of animals exposed to the cocaine-paired compartment (Lopes et al.,

17
2020). Therefore, endocannabinoid signalling possibly modulates drug-associated

memories through inhibition of hippocampal activation in presence of contextual cues.

Evidence from opposite roles of cannabinoid receptors also came from results showing

CB2 antagonists preventing the effects of CB1 antagonist on cocaine-induced CPP

(Gobira et al., 2019). Moreover, CB2 agonists also prevented acquisition and expression

of ethanol-induced CPP (Al Mansouri et al., 2014,Powers et al., 2015,Martín-Sánchez et

al., 2019). Accordingly, there is a reduction on CB2 gene expression in the ventral

midbrain of mice after prolonged alcohol consumption (Onaivi et al., 2008a). Other

studies, however, reached the opposite conclusions, since CB2 antagonism was able to

block acquisition of nicotine-induced CPP (Ignatowska-Jankowska et al.,

2013,Navarrete et al., 2013) and expression of alcohol-induced CPP (Martín-Sánchez et

al., 2019). This impairment may be result of aversive proprieties of CB2 antagonists,

since they did induce CPA (Navarrete et al., 2013).

Results from genetically modified animals have been mixed. Ethanol-induced CPP was

increased in CB2 knockout mice, although this phenotype was not recapitulated by

pharmacological interventions in the same study (Powers et al., 2015). Another study

also found that not only CPP, but also other behavioural responses to ethanol, were

exacerbated in these animals (Ortega-Álvaro et al., 2015). The opposite was found in

studies with nicotine, in which CB2 receptor deletion abolished CPP (Ignatowska-

Jankowska et al., 2013,Navarrete et al., 2013). Selective deletion of CB2 in

dopaminergic neurons also prevented CPP for nicotine (Canseco-Alba et al., 2019) and

ethanol (Canseco-Alba et al., 2018), although the effects of psychostimulant drugs such

as cocaine, amphetamine, and methamphetamine, remained unchanged (Canseco-Alba

et al., 2019). This discrepancy may be attributed to different drug actions on

dopaminergic singling in the mesolimbic pathway. While psychostimulants induce

18
direct increase of dopamine levels, nicotine acts indirectly on the dopaminergic system.

Finally, ~transgenic mice overexpressing CB2 receptors showed CPA, rather than CPP,

after cocaine administration (Aracil-Fernández et al., 2012).

Therefore, it still premature to draw any firm conclusion on the role of the CB2 receptor

on CPP. Based on the few pharmacological and genetic studies available so far, the

functions of CB2 receptor tend to be the opposite to those ascribed for CB1, at least for

cocaine-induced CPP. It is the activation of CB2, rather than its antagonism, that tend to

inhibit cocaine effects, although this observation does not necessary apply to the effects

of other drugs.

Endocannabinoid uptake and hydrolysis

Inhibition of endocannabinoid uptake (membrane transport) and hydrolysis represents

an interesting pharmacological approach to investigate the biological functions of

endocannabinoids. The specific roles of anandamide and 2-AG can be investigated

through the inhibition of their metabolizing enzymes, FAAH and MAGL, respectively.

Since endocannabinoids are produced on-demand, it is possible to selectively increase

their endogenous levels with temporal and anatomical resolution in the brain.

Systemic anandamide administration did not induce CPP (Scherma et al.,

2008a,Méndez-Díaz et al., 2012) or CPA per se (Scherma et al., 2008a). The same was

observed after increase in endogenous anandamide levels by inhibition of its reuptake or

hydrolysis (Gobbi et al., 2005,Bortolato et al., 2006,Scherma et al., 2008a,Scherma et

al., 2008b,Gamaleddin et al., 2013,DeVuono et al., 2017). However, when animals were

pretreated with a FAAH inhibitor, anandamide induced CPA, which was prevented by

19
CB1 antagonism (Scherma et al., 2008a). In fact, intravenous administration of

anandamide produced an increase in extracellular dopamine levels in NAcc shell, which

was magnified by FAAH inhibition and blocked by CB1 antagonism (Solinas et al.,

2006). Therefore, anandamide modulation of brain dopaminergic pathway results in

biphasic behavioural responses.

Although pharmacological FAAH inhibition does not serve as an unconditioned

stimulus to induce CPP, increasing anandamide levels modulates the effects of addictive

drugs in different phases of CPP. The main evidence has been obtained from studies

with nicotine. Inhibition of anandamide transport or hydrolysis impaired acquisition and

reinstatement of nicotine-induced CPP as well as nicotine-induced increase in dopamine

levels in the NAcc (Scherma et al., 2008b,Scherma et al., 2012). In contrast, another

study observed enhanced expression of nicotine-induced CPP after FAAH inhibition

(Merritt et al., 2008). Regarding morphine-induced CPP, FAAH and MAGL inhibition

did not affect CPA induced by morphine withdrawal (Gamage et al., 2015). One study

found changes in expression of mRNA levels of FAAH and MAGL in dorsal

hippocampus of animals submitted to different phases of morphine-induced CPP,

indicating alterations in the mechanisms of clearance of endocannabinoids following

expression and reinstatement (Li et al., 2017).

Some studies have investigated the phenotypes of FAAH knockout mice in CPP

protocols. An otherwise inactive dose of nicotine was able to induce CPP in these

animals through CB1 receptor facilitation, suggesting an endocannabinoid-mediated

enhancement of nicotine reward (Merritt et al., 2008,Pavon et al., 2018). There was also

an increase in dopamine levels in NAcc of FAAH knockout animals or after FAAH

pharmacological inhibition in wildtype animals (Pavon et al., 2018). Genetic deletion of

20
FAAH, however, failed to modify ethanol-induced CPP (Blednov et al., 2007).

Collected brain samples from animals submitted to ethanol-induced CPP found

decreased anandamide levels in the medial prefrontal cortex and ventral midbrain

(Martín-Sánchez et al., 2019). Possibly, repeated alcohol exposure misbalances

endocannabinoid levels throughout the mesocorticolimbic pathway, resulting in

increased dopaminergic activity.

Thus, the lack of effect of inhibitors of endocannabinoid membrane transport or

hydrolysis in place conditioning protocols implies that these compounds can be

developed as therapeutic agents devoid of rewarding properties and therefore with low

abuse liability. This is an important observation, since these compounds have been

investigated for the treatment of pain and various neuro-psychiatric disorders (Fowler,

2015a,b). Their effects against drug-induced CPP, however, are inconsistent, possibly

due to the opposite roles of CB1 and CB2 receptors in modulating drug rewarding

effects.

GENERAL DISCUSSION

Place conditioning protocols (CPP/CPA) have been instrumental to investigate the

effects of cannabinoids and the role of the endocannabinoid system on reward memory

associated with drugs of abuse. These studies contribute to the understanding of

important questions and hypothesis about cannabinoid addiction, including their impact

on the effects of other drugs, their biphasic effects on reward/aversion and their variable

effects on the first drug experience. Moreover, CPP studies have shed light on how

21
specific components of the endocannabinoid system relate to the biological effects of

other drugs, impacting the developing of new treatments for addiction.

THC and selective CB1 agonists induce CPP or CPA, depending on the dose and

previous experiences. The pattern emerging from studies investigating cannabinoids on

CPP (that is, the inconsistent and biphasic profiles) is similar to those observed with

other types of behavioural responses. It has also been challenging to detect the

reinforcing properties of THC in self-administration protocols (Justinova et al., 2005).

Moreover, in animal models of anxiety/fear, these cannabinoids tend to induce

anxiolytic-like effects at low doses, whereas the opposite generally occurs at high doses

(Moreira and Wotjak, 2010). This complex pattern of effects may result from

cannabinoid modulation of diverse neurotransmitter systems and effects on specific

neural subpopulations in certain brain regions (Lutz et al., 2015,Patel et al., 2017).

Accordingly, in humans, the effects of cannabis also change depending on dose and

previous stress experiences (Viveros et al., 2005). Contrary to THC and synthetic

cannabinoids, CBD induces neither reward nor aversion, even though it inhibits the

effects of other drugs in different phases of CPP, an interesting profile for therapeutic

exploitation. Noteworthy, CBD reduces cue-induced craving in humans with heroin use

disorder (Hurd et al., 2019), further supporting its potential therapeutic use in drug

addiction.

The potential role of CB1 receptor in the rewarding and reinforcing effects of various

drugs has drawn much attention to selective antagonists/inverse agonists as potential

treatments for addiction. Therefore, these compounds have been investigated in drug-

induced CPP. Various studies converge showing CB1 receptor antagonists reducing the

acquisition and expression of CPP induced by cocaine, alcohol, opioids and nicotine.

22
These consistent results might be explained by antagonism of CB1 receptor located in

neuronal terminals projecting onto dopaminergic neurons in VTA and NAcc. In these

regions, drugs of abuse recruits endocannabinoids, whereas CB1 receptor blockade

restrains the mesolimbic reward pathway (Cheer et al., 2007,Hernandez et al.,

2014,Wang et al., 2015), as illustrated in Figure 3. In addition, CB1 receptor in

hippocampus, amygdala, and prefrontal cortex possibly participate in the mechanisms

through which conditioned contextual cues trigger compulsive drug seeking (Moreira et

al., 2015) as well as control extinction and reinstatement of reward memories (Stern et

al., 2018,Goode and Maren, 2019). As result, and further reinforcing the translational

value of CPP, the CB1 antagonists/inverse agonist, rimonabant, surinabant and

taranabant have been investigated in humans studies for alcohol, nicotine and opioid use

disorders (Sloan et al., 2017,Chye et al., 2019). The expectations, however, have been

dampened by the psychiatric side effects of these compounds, which can facilitate

anxiety, panic and depressive mood, as revealed by several preclinical studies and by

the short-living experience with rimonabant for the treatment of obesity (Moreira and

Crippa, 2009) Since drug addiction is frequently co-morbid with other psychiatric

disorders (Verheul, 2001,Morozova et al., 2015,Rahman, 2015,Hillemacher and

Frieling, 2019), alternative possibilities have been pursued. One of them is the use of

neutral antagonists, which block the receptor without inverse agonistic activity and are

less prone to induce aversive side effects (Gueye et al., 2016).

More recently, the focus has shifted to the CB2 receptor. Notwithstanding the

controversies concerning its expression and function in brain, CB2 selective agonists

impair CPP induced by cocaine or ethanol. Two hypotheses, not mutually exclusive,

could explain CB2 receptor-modulation of drug reward. The non-neuronal hypothesis

can be built based on evidence that microglia and astrocytes, which express CB2

23
receptors (Stella, 2010), modulate mesolimbic dopamine signalling and drug reward

(Neuhofer and Kalivas, 2018,Linker et al., 2019,Stellwagen et al., 2019). Thus, CB2

agonists could inhibit glial function, including cytokine signalling, and indirectly

modulate neuronal activity related to drug reward. An alternative, neuronal hypothesis,

can also be posited. Evidence has emerged suggesting CB2 receptor expression in

central neurons, although in different locations from CB1. While the latter is expressed

in neuronal terminals projecting to dopaminergic neurons, CB2 is proposed to be located

directly in dopaminergic cell bodies in VTA, where its activation inhibits the release of

dopamine on NAcc in mice (Zhang et al., 2017). These hypothesis are illustrate in

Figure 3. CB2 is also expressed in the hippocampus (Stempel et al., 2016), a key

structure involved in CPP expression due its implication on spatial memory recovery

(Sjulson et al., 2018). Indeed, CB2 activation prevented neuronal activation in the

hippocampus of animals exposed to cocaine-induced CPP (Lopes et al., 2020). A

potential advantage of exploiting the favourable effects of CB2 receptor agonists is the

possibility of reducing the reward actions of drug of abuse without inducing the

psychiatric side effects associated with CB1 antagonists. Actually, CB2 receptor agonists

even alleviate anxiety-like behaviour in rodents (Bahi et al., 2014). Therefore, CB2 is a

promising target for new addiction treatments. However, CB2 involvement in drug

reward warrants further investigation, including studies concerning its expression and

function across different species. An approach deserving more evaluation is the

simultaneous modulation of CB1 and CB2 receptors, since recent findings suggest they

work in concert, with opposite function, to counteract reward responses (Gobira et al.,

2019).

The opposite roles of CB1 and CB2 may at least partially explain the inconsistent effects

of endocannabinoids hydrolysis inhibitors on CPP induced by drugs of abuse. Once

24
endocannabinoids levels are increased, they can promote simultaneous activation of

CB1 and CB2, despite differences on affinity. In addition, given the interaction between

endocannabinoid system and both inhibitory and excitatory neurotransmitters (Figure

3), the contradictory actions of endocannabinoid hydrolysis inhibitors are not surprising.

CONCLUSIONS

CPP is a valuable protocol for investigating the effects of compounds on every stage of

memories related to drug reward, i.e. acquisition, evocation, extinction, reinstatement,

and reconsolidation. It has been instrumental for deepening our understanding on the

rewarding and aversive properties of cannabinoids as well as the effects of substances

targeting the endocannabinoid system on contextual conditioning induced by various

drugs of abuse. “Neutral” CB1 receptor antagonists, selective CB2 receptor agonists and

substances combining both mechanisms emerge as therapeutic possibilities. Along with

other protocols, particularly sensitization and drug self-administration, place

conditioning studies point to the endocannabinoid system as a “drugable” target to

interfere with contextual memories responsible for initiating and maintaining drug

addiction.

25
Figure legends

Figure 1. The place conditioning protocol. In the pre-conditioning phase, animals (mice

or rats) are exposed to the apparatus with two distinguishable compartments, which they

are free to explore (in an unbiased protocol, animals spend similar times in both sides of

the box). In the conditioning phase, the drug is paired to one side of the box, where the

animals remain confined, and vehicle is paired to the opposite side. Finally, in the test

phase, drug-free animals explore all the compartments of the apparatus, similarly to the

pre-conditioning phase. An increase exploratory activity in the drug-paired side

indicates conditioned place preference (CPP), whereas an increase in time spent in the

vehicle-paired side indicates conditioned place aversion (CPA).

Figure 2. Schematic representation of the endocannabinoid system. Anandamide

(arachidonoyl ethanolamide, AEA) synthesis depends initially on glycerophospholipid

and phosphatidylethanolamine, which form N-arachidonoyl phosphatidylethanolamine

(NArPE) through N-acyltransferase. NArPE, in turn, is cleaved to anandamide through

N-acylphosphatidylethanolamine specific phospholipase D (PLD). 2-AG is formed by

glycerophospholipid cleavage facilitated by phospholipase C (PLC), followed by

diacylglycerol lipase (DAGL). They bind do CB1 and CB2 receptor and are internalised

by an endocannabinoid membrane transporter (EMT). Endocannabinoid main

hydrolysis processes depend on fatty acid amid hydrolase (FAAH), for anandamide, and

monoacylgycerol lipase (MAGL), for 2-AG.

26
Figure 3. Endocannabinoid modulation of the rewarding dopaminergic pathway. CB1

receptor could facilitate or inhibit dopaminergic activity and reward memory by

exerting inhibitory activity on GABAergic and glutamatergic terminals, respectively, in

the VTA/NAcc pathway. CB2 receptors could inhibit dopaminergic activity directly at

cell bodies at the VTA or through surround glial cells.

27
REFERENCES

Agmo A, Galvan A, Talamantes B (1995). Reward and reinforcement produced by

drinking sucrose: two processes that may depend on different neurotransmitters.

Pharmacol Biochem Behav 52(2):403-414. doi: 10.1016/0091-3057(95)00128-j.

Ahmad T, Laviolette SR (2017). Cannabinoid reward and aversion effects in the

posterior ventral tegmental area are mediated through dissociable opiate receptor

subtypes and separate amygdalar and accumbal dopamine receptor substrates.

Psychopharmacology (Berl) 234(15):2325-2336. doi: 10.1007/s00213-017-4669-7.

Al Mansouri S, Ojha S, Al Maamari E, Al Ameri M, Nurulain SM, Bahi A (2014). The

cannabinoid receptor 2 agonist, β-caryophyllene, reduced voluntary alcohol intake and

attenuated ethanol-induced place preference and sensitivity in mice. Pharmacol

Biochem Behav 124:260-268. doi: 10.1016/j.pbb.2014.06.025.

Aracil-Fernández A, Trigo JM, García-Gutiérrez MS, Ortega-Álvaro A, Ternianov A,

Navarro D et al. (2012). Decreased cocaine motor sensitization and self-administration

in mice overexpressing cannabinoid CB₂ receptors. Neuropsychopharmacology

37(7):1749-1763. doi: 10.1038/npp.2012.22.

Astur RS, Carew AW, Deaton BE (2014). Conditioned place preferences in humans

using virtual reality. Behav Brain Res 267:173-177. doi: 10.1016/j.bbr.2014.03.018.

Back SE, Hartwell K, DeSantis SM, Saladin M, McRae-Clark AL, Price KL et al.

(2010). Reactivity to laboratory stress provocation predicts relapse to cocaine. Drug

Alcohol Depend 106(1):21-27. doi: 10.1016/j.drugalcdep.2009.07.016.

28
Bahi A, Al Mansouri S, Al Memari E, Al Ameri M, Nurulain SM, Ojha S (2014). β-

Caryophyllene, a CB2 receptor agonist produces multiple behavioral changes relevant

to anxiety and depression in mice. Physiol Behav 135:119-124. doi:

10.1016/j.physbeh.2014.06.003.

Bardo MT, Bevins RA (2000). Conditioned place preference: what does it add to our

preclinical understanding of drug reward? Psychopharmacology (Berl) 153(1):31-43.

doi: 10.1007/s002130000569.

Bisogno T, Howell F, Williams G, Minassi A, Cascio MG, Ligresti A et al. (2003).

Cloning of the first sn1-DAG lipases points to the spatial and temporal regulation of

endocannabinoid signaling in the brain. J Cell Biol 163(3):463-468. doi:

10.1083/jcb.200305129.

Blednov YA, Cravatt BF, Boehm SL, Walker D, Harris RA (2007). Role of

endocannabinoids in alcohol consumption and intoxication: studies of mice lacking

fatty acid amide hydrolase. Neuropsychopharmacology 32(7):1570-1582. doi:

10.1038/sj.npp.1301274.

Bortolato M, Campolongo P, Mangieri RA, Scattoni ML, Frau R, Trezza V et al.

(2006). Anxiolytic-like properties of the anandamide transport inhibitor AM404.

Neuropsychopharmacology 31(12):2652-2659. doi: 10.1038/sj.npp.1301061.

Braida D, Pozzi M, Cavallini R, Sala M (2001). Conditioned place preference induced

by the cannabinoid agonist CP 55,940: interaction with the opioid system. Neuroscience

104(4):923-926. doi: 10.1016/s0306-4522(01)00210-x.

29
Braida D, Iosuè S, Pegorini S, Sala M (2004). Delta9-tetrahydrocannabinol-induced

conditioned place preference and intracerebroventricular self-administration in rats. Eur

J Pharmacol 506(1):63-69. doi: 10.1016/j.ejphar.2004.10.043.

Budzyńska B, Kruk M, Biała G (2009). Effects of the cannabinoid CB1 receptor

antagonist AM 251 on the reinstatement of nicotine-conditioned place preference by

drug priming in rats. Pharmacol Rep 61(2):304-310. doi: 10.1016/s1734-

1140(09)70036-2.

Canseco-Alba A, Schanz N, Ishiguro H, Liu QR, Onaivi ES (2018). Behavioral

Evaluation of Seeking and Preference of Alcohol in Mice Subjected to Stress. Bio

Protoc 8(20). doi: 10.21769/BioProtoc.3061.

Canseco-Alba A, Schanz N, Sanabria B, Zhao J, Lin Z, Liu QR et al. (2019). Behavioral

effects of psychostimulants in mutant mice with cell-type specific deletion of CB2

cannabinoid receptors in dopamine neurons. Behav Brain Res 360:286-297. doi:

10.1016/j.bbr.2018.11.043.

Castañé A, Valjent E, Ledent C, Parmentier M, Maldonado R, Valverde O (2002). Lack

of CB1 cannabinoid receptors modifies nicotine behavioural responses, but not nicotine

abstinence. Neuropharmacology 43(5):857-867. doi: 10.1016/s0028-3908(02)00118-1.

Chaperon F, Soubrié P, Puech AJ, Thiébot MH (1998). Involvement of central

cannabinoid (CB1) receptors in the establishment of place conditioning in rats.

Psychopharmacology (Berl) 135(4):324-332. doi: 10.1007/s002130050518.

30
Cheer JF, Kendall DA, Marsden CA (2000). Cannabinoid receptors and reward in the

rat: a conditioned place preference study. Psychopharmacology (Berl) 151(1):25-30.

doi: 10.1007/s002130000481.

Cheer JF, Wassum KM, Sombers LA, Heien ML, Ariansen JL, Aragona BJ et al.

(2007). Phasic dopamine release evoked by abused substances requires cannabinoid

receptor activation. J Neurosci 27(4):791-795. doi: 10.1523/JNEUROSCI.4152-

06.2007.

Childs E, de Wit H (2009). Amphetamine-induced place preference in humans. Biol

Psychiatry 65(10):900-904. doi: 10.1016/j.biopsych.2008.11.016.

Childs E, de Wit H (2013). Contextual conditioning enhances the psychostimulant and

incentive properties of d-amphetamine in humans. Addict Biol 18(6):985-992. doi:

10.1111/j.1369-1600.2011.00416.x.

Childs E, de Wit H (2016). Alcohol-induced place conditioning in moderate social

drinkers. Addiction 111(12):2157-2165. doi: 10.1111/add.13540.

Childs E, Astur RS, de Wit H (2017). Virtual reality conditioned place preference using

monetary reward. Behav Brain Res 322(Pt A):110-114. doi: 10.1016/j.bbr.2017.01.019.

Chye Y, Christensen E, Solowij N, Yücel M (2019). The Endocannabinoid System and

Cannabidiol's Promise for the Treatment of Substance Use Disorder. Front Psychiatry

10:63. doi: 10.3389/fpsyt.2019.00063.

Collaborators GAaDU (2018). The global burden of disease attributable to alcohol and

drug use in 195 countries and territories, 1990-2016: a systematic analysis for the

31
Global Burden of Disease Study 2016. Lancet Psychiatry 5(12):987-1012. doi:

10.1016/S2215-0366(18)30337-7.

Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gilula NB (1996).

Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid

amides. Nature 384(6604):83-87. doi: 10.1038/384083a0.

Cunningham CL, Gremel CM, Groblewski PA (2006). Drug-induced conditioned place

preference and aversion in mice. Nat Protoc 1(4):1662-1670. doi:

10.1038/nprot.2006.279.

Daza-Losada M, Miñarro J, Aguilar MA, Valverde O, Rodríguez-Arias M (2011).

Acute blockade of CB1 receptor leads to reinstatement of MDMA-induced conditioned

place preference. Pharmacol Biochem Behav 100(1):33-39. doi:

10.1016/j.pbb.2011.07.011.

de Carvalho CR, Takahashi RN (2017). Cannabidiol disrupts the reconsolidation of

contextual drug-associated memories in Wistar rats. Addict Biol 22(3):742-751. doi:

10.1111/adb.12366.

De Carvalho CR, Pamplona FA, Cruz JS, Takahashi RN (2014). Endocannabinoids

underlie reconsolidation of hedonic memories in Wistar rats. Psychopharmacology

(Berl) 231(7):1417-1425. doi: 10.1007/s00213-013-3331-2.

Delis F, Polissidis A, Poulia N, Justinova Z, Nomikos GG, Goldberg SR et al. (2017).

Attenuation of Cocaine-Induced Conditioned Place Preference and Motor Activity via

Cannabinoid CB2 Receptor Agonism and CB1 Receptor Antagonism in Rats. Int J

Neuropsychopharmacol 20(3):269-278. doi: 10.1093/ijnp/pyw102.

32
Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G et al. (1992).

Isolation and structure of a brain constituent that binds to the cannabinoid receptor.

Science 258(5090):1946-1949. doi: 10.1126/science.1470919.

DeVuono MV, Wills KL, MacPherson DV, Hrelja KM, Parker LA (2017). Effect of

footshock stress on place conditioning produced by Δ 9 -tetrahydrocannabinol and the

fatty acid amide hydrolase (FAAH) inhibitor, URB597, in Sprague-Dawley rats.

Psychopharmacology (Berl) 234(21):3229-3240. doi: 10.1007/s00213-017-4714-6.

Di Chiara G, Imperato A (1988). Drugs abused by humans preferentially increase

synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc

Natl Acad Sci U S A 85(14):5274-5278.

Dinh TP, Carpenter D, Leslie FM, Freund TF, Katona I, Sensi SL et al. (2002). Brain

monoglyceride lipase participating in endocannabinoid inactivation. Proc Natl Acad Sci

U S A 99(16):10819-10824. doi: 10.1073/pnas.152334899.

Elphick MR, Egertová M (2001). The neurobiology and evolution of cannabinoid

signalling. Philos Trans R Soc Lond B Biol Sci 356(1407):381-408. doi:

10.1098/rstb.2000.0787.

Everitt BJ, Robbins TW (2005). Neural systems of reinforcement for drug addiction:

from actions to habits to compulsion. Nat Neurosci 8(11):1481-1489. doi:

10.1038/nn1579.

Fang Q, Li FQ, Li YQ, Xue YX, He YY, Liu JF et al. (2011). Cannabinoid CB1

receptor antagonist rimonabant disrupts nicotine reward-associated memory in rats.

Pharmacol Biochem Behav 99(4):738-742. doi: 10.1016/j.pbb.2011.06.019.

33
Forget B, Hamon M, Thiébot MH (2005). Cannabinoid CB1 receptors are involved in

motivational effects of nicotine in rats. Psychopharmacology (Berl) 181(4):722-734.

doi: 10.1007/s00213-005-0015-6.

Fowler CJ (2015a). The Potential of Inhibitors of Endocannabinoid Metabolism for

Drug Development: A Critical Review. Handb Exp Pharmacol 231:95-128. doi:

10.1007/978-3-319-20825-1_4.

Fowler CJ (2015b). The potential of inhibitors of endocannabinoid metabolism as

anxiolytic and antidepressive drugs--A practical view. Eur Neuropsychopharmacol

25(6):749-762. doi: 10.1016/j.euroneuro.2015.02.005.

Gamage TF, Ignatowska-Jankowska BM, Muldoon PP, Cravatt BF, Damaj MI,

Lichtman AH (2015). Differential effects of endocannabinoid catabolic inhibitors on

morphine withdrawal in mice. Drug Alcohol Depend 146:7-16. doi:

10.1016/j.drugalcdep.2014.11.015.

Gamaleddin I, Guranda M, Scherma M, Fratta W, Makriyannis A, Vadivel SK et al.

(2013). AM404 attenuates reinstatement of nicotine seeking induced by nicotine-

associated cues and nicotine priming but does not affect nicotine- and food-taking. J

Psychopharmacol 27(6):564-571. doi: 10.1177/0269881113477710

0269881113477710 [pii].

Gobbi G, Bambico FR, Mangieri R, Bortolato M, Campolongo P, Solinas M et al.

(2005). Antidepressant-like activity and modulation of brain monoaminergic

transmission by blockade of anandamide hydrolysis. Proc Natl Acad Sci U S A

102(51):18620-18625. doi: 10.1073/pnas.0509591102.

34
Gobira PH, Oliveira AC, Gomes JS, da Silveira VT, Asth L, Bastos JR et al. (2019).

Opposing roles of CB1 and CB2 cannabinoid receptors in the stimulant and rewarding

effects of cocaine. Br J Pharmacol 176(10):1541-1551. doi: 10.1111/bph.14473.

Gong JP, Onaivi ES, Ishiguro H, Liu QR, Tagliaferro PA, Brusco A et al. (2006).

Cannabinoid CB2 receptors: immunohistochemical localization in rat brain. Brain Res

1071(1):10-23. doi: 10.1016/j.brainres.2005.11.035.

Goode TD, Maren S (2019). Common neurocircuitry mediating drug and fear relapse in

preclinical models. Psychopharmacology (Berl) 236(1):415-437. doi: 10.1007/s00213-

018-5024-3.

Gueye AB, Pryslawsky Y, Trigo JM, Poulia N, Delis F, Antoniou K et al. (2016). The

CB1 Neutral Antagonist AM4113 Retains the Therapeutic Efficacy of the Inverse

Agonist Rimonabant for Nicotine Dependence and Weight Loss with Better Psychiatric

Tolerability. Int J Neuropsychopharmacol 19(12). doi: 10.1093/ijnp/pyw068.

Hempel BJ, Wakeford AG, Clasen MM, Friar MA, Riley AL (2016). Delta-9-

tetrahydrocannabinol (THC) history fails to affect THC's ability to induce place

preferences in rats. Pharmacol Biochem Behav 144:1-6. doi:

10.1016/j.pbb.2016.02.007.

Herkenham M, Lynn AB, Johnson MR, Melvin LS, de Costa BR, Rice KC (1991).

Characterization and localization of cannabinoid receptors in rat brain: a quantitative in

vitro autoradiographic study. J Neurosci 11(2):563-583.

Hernandez G, Oleson EB, Gentry RN, Abbas Z, Bernstein DL, Arvanitogiannis A et al.

(2014). Endocannabinoids promote cocaine-induced impulsivity and its rapid

35
dopaminergic correlates. Biol Psychiatry 75(6):487-498. doi:

10.1016/j.biopsych.2013.09.005

S0006-3223(13)00824-X [pii].

Higley AE, Crane NA, Spadoni AD, Quello SB, Goodell V, Mason BJ (2011). Craving

in response to stress induction in a human laboratory paradigm predicts treatment

outcome in alcohol-dependent individuals. Psychopharmacology (Berl) 218(1):121-129.

doi: 10.1007/s00213-011-2355-8.

Hillemacher T, Frieling H (2019). Pharmacotherapeutic options for co-morbid

depression and alcohol dependence. Expert Opin Pharmacother 20(5):547-569. doi:

10.1080/14656566.2018.1561870.

Houchi H, Babovic D, Pierrefiche O, Ledent C, Daoust M, Naassila M (2005). CB1

receptor knockout mice display reduced ethanol-induced conditioned place preference

and increased striatal dopamine D2 receptors. Neuropsychopharmacology 30(2):339-

349. doi: 10.1038/sj.npp.1300568.

Hurd YL, Spriggs S, Alishayev J, Winkel G, Gurgov K, Kudrich C et al. (2019).

Cannabidiol for the Reduction of Cue-Induced Craving and Anxiety in Drug-Abstinent

Individuals With Heroin Use Disorder: A Double-Blind Randomized Placebo-

Controlled Trial. Am J Psychiatry 176(11):911-922. doi:

10.1176/appi.ajp.2019.18101191.

Huston JP, Silva MA, Topic B, Müller CP (2013). What's conditioned in conditioned

place preference? Trends Pharmacol Sci 34(3):162-166. doi:

10.1016/j.tips.2013.01.004.

36
Hutcheson DM, Tzavara ET, Smadja C, Valjent E, Roques BP, Hanoune J et al. (1998).

Behavioural and biochemical evidence for signs of abstinence in mice chronically

treated with delta-9-tetrahydrocannabinol. Br J Pharmacol 125(7):1567-1577. doi:

10.1038/sj.bjp.0702228.

Hyatt WS, Fantegrossi WE (2014). Δ9-THC exposure attenuates aversive effects and

reveals appetitive effects of K2/'Spice' constituent JWH-018 in mice. Behav Pharmacol

25(3):253-257. doi: 10.1097/FBP.0000000000000034.

Ignatowska-Jankowska BM, Muldoon PP, Lichtman AH, Damaj MI (2013). The

cannabinoid CB2 receptor is necessary for nicotine-conditioned place preference, but

not other behavioral effects of nicotine in mice. Psychopharmacology (Berl)

229(4):591-601. doi: 10.1007/s00213-013-3117-6.

Izzo AA, Borrelli F, Capasso R, Di Marzo V, Mechoulam R (2009). Non-psychotropic

plant cannabinoids: new therapeutic opportunities from an ancient herb. Trends

Pharmacol Sci 30(10):515-527. doi: 10.1016/j.tips.2009.07.006.

Jardinaud F, Roques BP, Noble F (2006). Tolerance to the reinforcing effects of

morphine in delta9-tetrahydrocannabinol treated mice. Behav Brain Res 173(2):255-

261. doi: 10.1016/j.bbr.2006.06.027.

Justinova Z, Goldberg SR, Heishman SJ, Tanda G (2005). Self-administration of

cannabinoids by experimental animals and human marijuana smokers. Pharmacol

Biochem Behav 81(2):285-299. doi: S0091-3057(05)00139-5 [pii]

10.1016/j.pbb.2005.01.026.

37
Karimi S, Azizi P, Shamsizadeh A, Haghparast A (2013). Role of intra-accumbal

cannabinoid CB1 receptors in the potentiation, acquisition and expression of morphine-

induced conditioned place preference. Behav Brain Res 247:125-131. doi:

10.1016/j.bbr.2013.03.022.

Katz RJ, Gormezano G (1979). A rapid and inexpensive technique for assessing the

reinforcing effects of opiate drugs. Pharmacol Biochem Behav 11(2):231-233. doi:

10.1016/0091-3057(79)90019-4.

Koob GF, Le Moal M (2008). Review. Neurobiological mechanisms for opponent

motivational processes in addiction. Philos Trans R Soc Lond B Biol Sci

363(1507):3113-3123. doi: 10.1098/rstb.2008.0094.

Koob GF, Volkow ND (2016). Neurobiology of addiction: a neurocircuitry analysis.

Lancet Psychiatry 3(8):760-773. doi: 10.1016/S2215-0366(16)00104-8.

Kreitzer AC, Regehr WG (2001). Retrograde inhibition of presynaptic calcium influx

by endogenous cannabinoids at excitatory synapses onto Purkinje cells. Neuron

29(3):717-727. doi: 10.1016/s0896-6273(01)00246-x.

Kubilius RA, Kaplick PM, Wotjak CT (2018). Highway to hell or magic smoke? The

dose-dependence of Δ9-THC in place conditioning paradigms. Learn Mem 25(9):446-

454. doi: 10.1101/lm.046870.117.

Kuhn BN, Kalivas PW, Bobadilla AC (2019). Understanding Addiction Using Animal

Models. Front Behav Neurosci 13:262. doi: 10.3389/fnbeh.2019.00262.

38
Le Foll B, Goldberg SR (2004). Rimonabant, a CB1 antagonist, blocks nicotine-

conditioned place preferences. Neuroreport 15(13):2139-2143. doi: 10.1097/00001756-

200409150-00028.

Le Foll B, Wiggins M, Goldberg SR (2006). Nicotine pre-exposure does not potentiate

the locomotor or rewarding effects of Delta-9-tetrahydrocannabinol in rats. Behav

Pharmacol 17(2):195-199. doi: 10.1097/01.fbp.0000197460.16516.81.

Le Foll B, Forget B, Aubin HJ, Goldberg SR (2008). Blocking cannabinoid CB1

receptors for the treatment of nicotine dependence: insights from pre-clinical and

clinical studies. Addict Biol 13(2):239-252. doi: 10.1111/j.1369-1600.2008.00113.x.

LeGates TA, Kvarta MD, Tooley JR, Francis TC, Lobo MK, Creed MC et al. (2018).

Reward behaviour is regulated by the strength of hippocampus-nucleus accumbens

synapses. Nature 564(7735):258-262. doi: 10.1038/s41586-018-0740-8.

Li W, Zhang CL, Qiu ZG (2017). Differential expression of endocannabinoid system-

related genes in the dorsal hippocampus following expression and reinstatement of

morphine conditioned place preference in mice. Neurosci Lett 643:38-44. doi:

10.1016/j.neulet.2017.02.025.

Linker KE, Cross SJ, Leslie FM (2019). Glial mechanisms underlying substance use

disorders. Eur J Neurosci 50(3):2574-2589. doi: 10.1111/ejn.14163.

Liu Y, Le Foll B, Wang X, Lu L (2008). Conditioned place preference induced by licit

drugs: establishment, extinction, and reinstatement. ScientificWorldJournal 8:1228-

1245. doi: 10.1100/tsw.2008.154.

39
Lopes JB, Bastos JR, Costa RB, Aguiar DC, Moreira FA (2020). The roles of

cannabinoid CB1 and CB2 receptors in cocaine-induced behavioral sensitization and

conditioned place preference in mice. Psychopharmacology (Berl) 237(2):385-394. doi:

10.1007/s00213-019-05370-5.

Luján M, Castro-Zavala A, Alegre-Zurano L, Valverde O (2018). Repeated Cannabidiol

treatment reduces cocaine intake and modulates neural proliferation and CB1R

expression in the mouse hippocampus. Neuropharmacology 143:163-175. doi:

10.1016/j.neuropharm.2018.09.043.

Lutz B, Marsicano G, Maldonado R, Hillard CJ (2015). The endocannabinoid system in

guarding against fear, anxiety and stress. Nat Rev Neurosci 16(12):705-718. doi:

10.1038/nrn4036.

Lynskey MT, Heath AC, Bucholz KK, Slutske WS, Madden PA, Nelson EC et al.

(2003). Escalation of drug use in early-onset cannabis users vs co-twin controls. JAMA

289(4):427-433. doi: 10.1001/jama.289.4.427.

López-Moreno JA, González-Cuevas G, Rodríguez de Fonseca F, Navarro M (2004).

Long-lasting increase of alcohol relapse by the cannabinoid receptor agonist WIN

55,212-2 during alcohol deprivation. J Neurosci 24(38):8245-8252. doi:

10.1523/JNEUROSCI.2179-04.2004.

Mantsch JR, Baker DA, Funk D, Lê AD, Shaham Y (2016). Stress-Induced

Reinstatement of Drug Seeking: 20 Years of Progress. Neuropsychopharmacology

41(1):335-356. doi: 10.1038/npp.2015.142.

40
Manzanares J, Cabañero D, Puente N, García-Gutiérrez MS, Grandes P, Maldonado R

(2018). Role of the endocannabinoid system in drug addiction. Biochem Pharmacol

157:108-121. doi: 10.1016/j.bcp.2018.09.013.

Manzanedo C, Aguilar MA, Rodríguez-Arias M, Navarro M, Miñarro J (2004).

Cannabinoid agonist-induced sensitisation to morphine place preference in mice.

Neuroreport 15(8):1373-1377. doi: 10.1097/01.wnr.0000126217.87116.8c.

Marcus DJ, Henderson-Redmond AN, Gonek M, Zee ML, Farnsworth JC, Amin RA et

al. (2017). Mice expressing a "hyper-sensitive" form of the CB1 cannabinoid receptor

(CB1) show modestly enhanced alcohol preference and consumption. PLoS One

12(4):e0174826. doi: 10.1371/journal.pone.0174826.

Markos JR, Harris HM, Gul W, ElSohly MA, Sufka KJ (2018). Effects of Cannabidiol

on Morphine Conditioned Place Preference in Mice. Planta Med 84(4):221-224. doi:

10.1055/s-0043-117838.

Martin M, Ledent C, Parmentier M, Maldonado R, Valverde O (2000). Cocaine, but not

morphine, induces conditioned place preference and sensitization to locomotor

responses in CB1 knockout mice. Eur J Neurosci 12(11):4038-4046. doi:

10.1046/j.1460-9568.2000.00287.x.

Martín-Sánchez A, Warnault V, Montagud-Romero S, Pastor A, Mondragón N, De La

Torre R et al. (2019). Alcohol-induced conditioned place preference is modulated by

CB2 cannabinoid receptors and modifies levels of endocannabinoids in the

mesocorticolimbic system. Pharmacol Biochem Behav 183:22-31. doi:

10.1016/j.pbb.2019.06.007.

41
Mas-Nieto M, Pommier B, Tzavara ET, Caneparo A, Da Nascimento S, Le Fur G et al.

(2001). Reduction of opioid dependence by the CB(1) antagonist SR141716A in mice:

evaluation of the interest in pharmacotherapy of opioid addiction. Br J Pharmacol

132(8):1809-1816. doi: 10.1038/sj.bjp.0703990.

Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI (1990). Structure of a

cannabinoid receptor and functional expression of the cloned cDNA. Nature

346(6284):561-564. doi: 10.1038/346561a0.

Mechoulam R, Parker LA (2013). The endocannabinoid system and the brain. Annu

Rev Psychol 64:21-47. doi: 10.1146/annurev-psych-113011-143739.

Mechoulam R, Hanuš LO, Pertwee R, Howlett AC (2014). Early phytocannabinoid

chemistry to endocannabinoids and beyond. Nat Rev Neurosci 15(11):757-764. doi:

10.1038/nrn3811.

Merritt LL, Martin BR, Walters C, Lichtman AH, Damaj MI (2008). The endogenous

cannabinoid system modulates nicotine reward and dependence. J Pharmacol Exp Ther

326(2):483-492. doi: 10.1124/jpet.108.138321.

Milella MS, Fotros A, Gravel P, Casey KF, Larcher K, Verhaeghe JA et al. (2016).

Cocaine cue-induced dopamine release in the human prefrontal cortex. J Psychiatry

Neurosci 41(5):322-330. doi: 10.1503/jpn.150207.

Milton AL, Everitt BJ (2012). The persistence of maladaptive memory: addiction, drug

memories and anti-relapse treatments. Neurosci Biobehav Rev 36(4):1119-1139. doi:

10.1016/j.neubiorev.2012.01.002.

42
Molet M, Billiet G, Bardo MT (2013). Conditioned place preference and aversion for

music in a virtual reality environment. Behav Processes 92:31-35. doi:

10.1016/j.beproc.2012.10.001.

Moreira FA, Crippa JA (2009). The psychiatric side-effects of rimonabant. Braz J

Psychiatry 31(2):145-153. doi: 10.1590/s1516-44462009000200012.

Moreira FA, Wotjak CT (2010). Cannabinoids and anxiety. Curr Top Behav Neurosci

2:429-450. doi: 10.1007/7854_2009_16.

Moreira FA, Jupp B, Belin D, Dalley JW (2015). Endocannabinoids and striatal

function: implications for addiction-related behaviours. Behav Pharmacol 26(1-2):59-

72. doi: 10.1097/FBP.0000000000000109.

Morgan CJ, Das RK, Joye A, Curran HV, Kamboj SK (2013). Cannabidiol reduces

cigarette consumption in tobacco smokers: preliminary findings. Addict Behav

38(9):2433-2436. doi: 10.1016/j.addbeh.2013.03.011.

Morozova M, Rabin RA, George TP (2015). Co-morbid tobacco use disorder and

depression: A re-evaluation of smoking cessation therapy in depressed smokers. Am J

Addict 24(8):687-694. doi: 10.1111/ajad.12277.

Munro S, Thomas KL, Abu-Shaar M (1993). Molecular characterization of a peripheral

receptor for cannabinoids. Nature 365(6441):61-65. doi: 10.1038/365061a0.

Mátyás F, Urbán GM, Watanabe M, Mackie K, Zimmer A, Freund TF et al. (2008).

Identification of the sites of 2-arachidonoylglycerol synthesis and action imply

retrograde endocannabinoid signaling at both GABAergic and glutamatergic synapses

43
in the ventral tegmental area. Neuropharmacology 54(1):95-107. doi:

10.1016/j.neuropharm.2007.05.028.

Méndez-Díaz M, Rueda-Orozco PE, Ruiz-Contreras AE, Prospéro-García O (2012).

The endocannabinoid system modulates the valence of the emotion associated to food

ingestion. Addict Biol 17(4):725-735. doi: 10.1111/j.1369-1600.2010.00271.x.

Navarrete F, Rodríguez-Arias M, Martín-García E, Navarro D, García-Gutiérrez MS,

Aguilar MA et al. (2013). Role of CB2 cannabinoid receptors in the rewarding,

reinforcing, and physical effects of nicotine. Neuropsychopharmacology 38(12):2515-

2524. doi: 10.1038/npp.2013.157.

Neuhofer D, Kalivas P (2018). Metaplasticity at the addicted tetrapartite synapse: A

common denominator of drug induced adaptations and potential treatment target for

addiction. Neurobiol Learn Mem 154:97-111. doi: 10.1016/j.nlm.2018.02.007.

O'Brien CP (2008). Review. Evidence-based treatments of addiction. Philos Trans R

Soc Lond B Biol Sci 363(1507):3277-3286. doi: 10.1098/rstb.2008.0105.

Ohno-Shosaku T, Maejima T, Kano M (2001). Endogenous cannabinoids mediate

retrograde signals from depolarized postsynaptic neurons to presynaptic terminals.

Neuron 29(3):729-738. doi: 10.1016/s0896-6273(01)00247-1.

Okamoto Y, Morishita J, Tsuboi K, Tonai T, Ueda N (2004). Molecular characterization

of a phospholipase D generating anandamide and its congeners. J Biol Chem

279(7):5298-5305. doi: 10.1074/jbc.M306642200.

44
Onaivi ES, Carpio O, Ishiguro H, Schanz N, Uhl GR, Benno R (2008a). Behavioral

effects of CB2 cannabinoid receptor activation and its influence on food and alcohol

consumption. Ann N Y Acad Sci 1139:426-433. doi: 10.1196/annals.1432.035.

Onaivi ES, Ishiguro H, Gong JP, Patel S, Meozzi PA, Myers L et al. (2008b). Brain

neuronal CB2 cannabinoid receptors in drug abuse and depression: from mice to human

subjects. PLoS One 3(2):e1640. doi: 10.1371/journal.pone.0001640.

Ortega-Álvaro A, Ternianov A, Aracil-Fernández A, Navarrete F, García-Gutiérrez MS,

Manzanares J (2015). Role of cannabinoid CB2 receptor in the reinforcing actions of

ethanol. Addict Biol 20(1):43-55. doi: 10.1111/adb.12076.

Parker LA, Mcdonald RV (2000). Reinstatement of both a conditioned place preference

and a conditioned place aversion with drug primes. Pharmacol Biochem Behav

66(3):559-561. doi: 10.1016/s0091-3057(00)00222-7.

Parker LA, Burton P, Sorge RE, Yakiwchuk C, Mechoulam R (2004). Effect of low

doses of delta9-tetrahydrocannabinol and cannabidiol on the extinction of cocaine-

induced and amphetamine-induced conditioned place preference learning in rats.

Psychopharmacology (Berl) 175(3):360-366. doi: 10.1007/s00213-004-1825-7.

Parsons LH, Hurd YL (2015). Endocannabinoid signalling in reward and addiction. Nat

Rev Neurosci 16(10):579-594. doi: 10.1038/nrn4004.

Patel S, Hill MN, Cheer JF, Wotjak CT, Holmes A (2017). The endocannabinoid

system as a target for novel anxiolytic drugs. Neurosci Biobehav Rev 76(Pt A):56-66.

doi: 10.1016/j.neubiorev.2016.12.033.

45
Pavon FJ, Serrano A, Sidhpura N, Polis I, Stouffer D, de Fonseca FR et al. (2018). Fatty

acid amide hydrolase (FAAH) inactivation confers enhanced sensitivity to nicotine-

induced dopamine release in the mouse nucleus accumbens. Addict Biol 23(2):723-734.

doi: 10.1111/adb.12531.

Pertwee RG, Howlett AC, Abood ME, Alexander SP, Di Marzo V, Elphick MR et al.

(2010). International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid

receptors and their ligands: beyond CB₁ and CB₂ . Pharmacol Rev 62(4):588-631. doi:

10.1124/pr.110.003004.

Polissidis A, Chouliara O, Galanopoulos A, Marselos M, Papadopoulou-Daifoti Z,

Antoniou K (2009). Behavioural and dopaminergic alterations induced by a low dose of

WIN 55,212-2 in a conditioned place preference procedure. Life Sci 85(5-6):248-254.

doi: 10.1016/j.lfs.2009.05.015.

Powers MS, Breit KR, Chester JA (2015). Genetic Versus Pharmacological Assessment

of the Role of Cannabinoid Type 2 Receptors in Alcohol Reward-Related Behaviors.

Alcohol Clin Exp Res 39(12):2438-2446. doi: 10.1111/acer.12894.

Quinn HR, Matsumoto I, Callaghan PD, Long LE, Arnold JC, Gunasekaran N et al.

(2008). Adolescent rats find repeated Delta(9)-THC less aversive than adult rats but

display greater residual cognitive deficits and changes in hippocampal protein

expression following exposure. Neuropsychopharmacology 33(5):1113-1126. doi:

10.1038/sj.npp.1301475.

Rahman S (2015). Targeting brain nicotinic acetylcholine receptors to treat major

depression and co-morbid alcohol or nicotine addiction. CNS Neurol Disord Drug

Targets 14(5):647-653. doi: 10.2174/1871527314666150429112954.

46
Ramiro-Fuentes S, Ortiz O, Moratalla R, Fernandez-Espejo E (2010). Intra-accumbens

rimonabant is rewarding but induces aversion to cocaine in cocaine-treated rats, as does

in vivo accumbal cannabinoid CB1 receptor silencing: critical role for glutamate

receptors. Neuroscience 167(2):205-215. doi: 10.1016/j.neuroscience.2010.02.019.

Rashidy-Pour A, Pahlevani P, Vaziri A, Shaigani P, Zarepour L, Vafaei AA et al.

(2013). Involvement of CB1 receptors in the ventral tegmental area in the potentiation

of morphine rewarding properties in acquisition but not expression in the conditioned

place preference model. Behav Brain Res 247:259-267. doi: 10.1016/j.bbr.2013.03.015.

Rodríguez-Arias M, Manzanedo C, Roger-Sánchez C, Do Couto BR, Aguilar MA,

Miñarro J (2010). Effect of adolescent exposure to WIN 55212-2 on the acquisition and

reinstatement of MDMA-induced conditioned place preference. Prog

Neuropsychopharmacol Biol Psychiatry 34(1):166-171. doi:

10.1016/j.pnpbp.2009.10.019.

Rodríguez-Arias M, Roger-Sánchez C, Vilanova I, Revert N, Manzanedo C, Miñarro J

et al. (2016). Effects of Cannabinoid Exposure during Adolescence on the Conditioned

Rewarding Effects of WIN 55212-2 and Cocaine in Mice: Influence of the Novelty-

Seeking Trait. Neural Plast 2016:6481862. doi: 10.1155/2016/6481862.

Sañudo-Peña MC, Tsou K, Delay ER, Hohman AG, Force M, Walker JM (1997).

Endogenous cannabinoids as an aversive or counter-rewarding system in the rat.

Neurosci Lett 223(2):125-128. doi: 10.1016/s0304-3940(97)13424-3.

Scherma M, Medalie J, Fratta W, Vadivel SK, Makriyannis A, Piomelli D et al.

(2008a). The endogenous cannabinoid anandamide has effects on motivation and

47
anxiety that are revealed by fatty acid amide hydrolase (FAAH) inhibition.

Neuropharmacology 54(1):129-140. doi: 10.1016/j.neuropharm.2007.08.011.

Scherma M, Justinová Z, Zanettini C, Panlilio LV, Mascia P, Fadda P et al. (2012). The

anandamide transport inhibitor AM404 reduces the rewarding effects of nicotine and

nicotine-induced dopamine elevations in the nucleus accumbens shell in rats. Br J

Pharmacol 165(8):2539-2548. doi: 10.1111/j.1476-5381.2011.01467.x.

Scherma M, Panlilio LV, Fadda P, Fattore L, Gamaleddin I, Le Foll B et al. (2008b).

Inhibition of anandamide hydrolysis by cyclohexyl carbamic acid 3'-carbamoyl-3-yl

ester (URB597) reverses abuse-related behavioral and neurochemical effects of nicotine

in rats. J Pharmacol Exp Ther 327(2):482-490. doi: 10.1124/jpet.108.142224.

Shinohara F, Kamii H, Minami M, Kaneda K (2017). The Role of Dopaminergic

Signaling in the Medial Prefrontal Cortex for the Expression of Cocaine-Induced

Conditioned Place Preference in Rats. Biol Pharm Bull 40(11):1983-1989. doi:

10.1248/bpb.b17-00614.

Singh ME, Verty AN, McGregor IS, Mallet PE (2004). A cannabinoid receptor

antagonist attenuates conditioned place preference but not behavioural sensitization to

morphine. Brain Res 1026(2):244-253. doi: 10.1016/j.brainres.2004.08.027.

Sinha R, Garcia M, Paliwal P, Kreek MJ, Rounsaville BJ (2006). Stress-induced

cocaine craving and hypothalamic-pituitary-adrenal responses are predictive of cocaine

relapse outcomes. Arch Gen Psychiatry 63(3):324-331. doi: 10.1001/archpsyc.63.3.324.

Sinha R, Fox HC, Hong KI, Hansen J, Tuit K, Kreek MJ (2011). Effects of adrenal

sensitivity, stress- and cue-induced craving, and anxiety on subsequent alcohol relapse

48
and treatment outcomes. Arch Gen Psychiatry 68(9):942-952. doi:

10.1001/archgenpsychiatry.2011.49.

Sjulson L, Peyrache A, Cumpelik A, Cassataro D, Buzsáki G (2018). Cocaine Place

Conditioning Strengthens Location-Specific Hippocampal Coupling to the Nucleus

Accumbens. Neuron 98(5):926-934.e925. doi: 10.1016/j.neuron.2018.04.015.

Sloan ME, Gowin JL, Ramchandani VA, Hurd YL, Le Foll B (2017). The

endocannabinoid system as a target for addiction treatment: Trials and tribulations.

Neuropharmacology 124:73-83. doi: 10.1016/j.neuropharm.2017.05.031.

Solinas M, Justinova Z, Goldberg SR, Tanda G (2006). Anandamide administration

alone and after inhibition of fatty acid amide hydrolase (FAAH) increases dopamine

levels in the nucleus accumbens shell in rats. J Neurochem 98(2):408-419. doi:

10.1111/j.1471-4159.2006.03880.x.

Stella N (2010). Cannabinoid and cannabinoid-like receptors in microglia, astrocytes,

and astrocytomas. Glia 58(9):1017-1030. doi: 10.1002/glia.20983.

Stellwagen D, Kemp GM, Valade S, Chambon J (2019). Glial regulation of synaptic

function in models of addiction. Curr Opin Neurobiol 57:179-185. doi:

10.1016/j.conb.2019.02.010.

Stempel AV, Stumpf A, Zhang HY, Özdoğan T, Pannasch U, Theis AK et al. (2016).

Cannabinoid Type 2 Receptors Mediate a Cell Type-Specific Plasticity in the

Hippocampus. Neuron 90(4):795-809. doi: 10.1016/j.neuron.2016.03.034.

Stern CAJ, de Carvalho CR, Bertoglio LJ, Takahashi RN (2018). Effects of

Cannabinoid Drugs on Aversive or Rewarding Drug-Associated Memory Extinction

49
and Reconsolidation. Neuroscience 370:62-80. doi:

10.1016/j.neuroscience.2017.07.018.

Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, Itoh K et al. (1995). 2-

Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain.

Biochem Biophys Res Commun 215(1):89-97. doi: 10.1006/bbrc.1995.2437.

Thanos PK, Dimitrakakis ES, Rice O, Gifford A, Volkow ND (2005). Ethanol self-

administration and ethanol conditioned place preference are reduced in mice lacking

cannabinoid CB1 receptors. Behav Brain Res 164(2):206-213. doi:

10.1016/j.bbr.2005.06.021.

Tsou K, Brown S, Sañudo-Peña MC, Mackie K, Walker JM (1998).

Immunohistochemical distribution of cannabinoid CB1 receptors in the rat central

nervous system. Neuroscience 83(2):393-411. doi: 10.1016/s0306-4522(97)00436-3.

Tzschentke TM (1998). Measuring reward with the conditioned place preference

paradigm: a comprehensive review of drug effects, recent progress and new issues. Prog

Neurobiol 56(6):613-672. doi: 10.1016/s0301-0082(98)00060-4.

Tzschentke TM (2007). Measuring reward with the conditioned place preference (CPP)

paradigm: update of the last decade. Addict Biol 12(3-4):227-462. doi: 10.1111/j.1369-

1600.2007.00070.x.

Uchigashima M, Narushima M, Fukaya M, Katona I, Kano M, Watanabe M (2007).

Subcellular arrangement of molecules for 2-arachidonoyl-glycerol-mediated retrograde

signaling and its physiological contribution to synaptic modulation in the striatum. J

Neurosci 27(14):3663-3676. doi: 10.1523/JNEUROSCI.0448-07.2007.

50
Valjent E, Maldonado R (2000). A behavioural model to reveal place preference to delta

9-tetrahydrocannabinol in mice. Psychopharmacology (Berl) 147(4):436-438. doi:

10.1007/s002130050013.

Valverde O, Noble F, Beslot F, Daugé V, Fournié-Zaluski MC, Roques BP (2001).

Delta9-tetrahydrocannabinol releases and facilitates the effects of endogenous

enkephalins: reduction in morphine withdrawal syndrome without change in rewarding

effect. Eur J Neurosci 13(9):1816-1824. doi: 10.1046/j.0953-816x.2001.01558.x.

Vann RE, Gamage TF, Warner JA, Marshall EM, Taylor NL, Martin BR et al. (2008).

Divergent effects of cannabidiol on the discriminative stimulus and place conditioning

effects of Delta(9)-tetrahydrocannabinol. Drug Alcohol Depend 94(1-3):191-198. doi:

10.1016/j.drugalcdep.2007.11.017.

Vaughn LK, Mantsch JR, Vranjkovic O, Stroh G, Lacourt M, Kreutter M et al. (2012).

Cannabinoid receptor involvement in stress-induced cocaine reinstatement: potential

interaction with noradrenergic pathways. Neuroscience 204:117-124. doi:

10.1016/j.neuroscience.2011.08.021.

Verheul R (2001). Co-morbidity of personality disorders in individuals with substance

use disorders. Eur Psychiatry 16(5):274-282. doi: 10.1016/s0924-9338(01)00578-8.

Viveros MP, Marco EM, File SE (2005). Endocannabinoid system and stress and

anxiety responses. Pharmacol Biochem Behav 81(2):331-342. doi:

10.1016/j.pbb.2005.01.029.

Vlachou S, Nomikos GG, Stephens DN, Panagis G (2007). Lack of evidence for

appetitive effects of Delta 9-tetrahydrocannabinol in the intracranial self-stimulation

51
and conditioned place preference procedures in rodents. Behav Pharmacol 18(4):311-

319. doi: 10.1097/FBP.0b013e3282186cf2.

Wang H, Treadway T, Covey DP, Cheer JF, Lupica CR (2015). Cocaine-Induced

Endocannabinoid Mobilization in the Ventral Tegmental Area. Cell Rep 12(12):1997-

2008. doi: 10.1016/j.celrep.2015.08.041.

Welberg L (2011). Addiction: from mechanisms to treatment. Nat Rev Neurosci

12(11):621. doi: 10.1038/nrn3131.

Wilson RI, Nicoll RA (2001). Endogenous cannabinoids mediate retrograde signalling

at hippocampal synapses. Nature 410(6828):588-592. doi: 10.1038/35069076.

Wise RA, Koob GF (2014). The development and maintenance of drug addiction.

Neuropsychopharmacology 39(2):254-262. doi: 10.1038/npp.2013.261.

Xi ZX, Peng XQ, Li X, Song R, Zhang HY, Liu QR et al. (2011). Brain cannabinoid

CB(2) receptors modulate cocaine's actions in mice. Nat Neurosci 14(9):1160-1166.

doi: 10.1038/nn.2874

nn.2874 [pii].

Yu LL, Zhou SJ, Wang XY, Liu JF, Xue YX, Jiang W et al. (2011). Effects of

cannabinoid CB₁ receptor antagonist rimonabant on acquisition and reinstatement of

psychostimulant reward memory in mice. Behav Brain Res 217(1):111-116. doi:

10.1016/j.bbr.2010.10.008.

Zangen A, Solinas M, Ikemoto S, Goldberg SR, Wise RA (2006). Two brain sites for

cannabinoid reward. J Neurosci 26(18):4901-4907. doi: 26/18/4901 [pii]

52
10.1523/JNEUROSCI.3554-05.2006.

Zhang HY, Gao M, Liu QR, Bi GH, Li X, Yang HJ et al. (2014). Cannabinoid CB2

receptors modulate midbrain dopamine neuronal activity and dopamine-related behavior

in mice. Proc Natl Acad Sci U S A 111(46):E5007-5015. doi:

10.1073/pnas.1413210111.

Zhang HY, Gao M, Shen H, Bi GH, Yang HJ, Liu QR et al. (2017). Expression of

functional cannabinoid CB 2 receptor in VTA dopamine neurons in rats. Addict Biol

22(3):752-765. doi: 10.1111/adb.12367.

53
Table

Tables

Table 1: Selected cannabinoids and compounds targeting the endocannabinoid system.

Compound Mechanism

Phytocannabinoids

THC Partial agonist at CB1 and CB2

CBD Various mechanisms

Synthetic cannabinoids

CP-55940 CB1/CB2 agonist

HU210 CB1/CB2 agonist

WIN-55212-2 CB1/CB2 agonist

ACEA CB1 agonist

JWH133 CB2 agonist

Antagonists/inverse agonists

AM251 CB1 antagonist/inverse agonist

Rimonabant (SR 141716) CB1 antagonist/inverse agonist

AM4113 CB1 “neutral” antagonist

AM630 CB2 antagonist

Endocannabinoids

Anandamide CB1/CB2 agonist

2-arachidonoylglycerol (2-AG) CB1/CB2 agonist

Endocannabinoid hydrolysis and uptake inhibitors

URB597 FAAH inhibitor

JZL184 MAGL inhibitor


AM404 Endocannabinoid uptake inhibitor
Figure Click here to download Figure Figure CPP.tiff
Figure Click here to download Figure Figure eCBs.tiff
Figure Click here to download Figure Figure pathway SIMPLES.tiff

You might also like