ArticlePDF Available

Role of Neuronal Nicotinic Acetylcholine Receptors in Cannabinoid Dependence

Authors:

Abstract

Cannabis is among the most widely consumed psychoactive drugs around the world and cannabis use disorder (CUD) has no current approved pharmacological treatment. Nicotine and cannabis are commonly co-used which suggests there to be overlapping neurobiological actions supported primarily by the co-distribution of both receptor systems in the brain. There appears to be strong rationale to explore the role that nicotinic receptors play in cannabinoid dependence. Preclinical studies suggest that the ɑ7 nAChR subtype may play a role in modulating the reinforcing and discriminative stimulus effects of cannabinoids, while the ɑ4β2* nAChR subtype may be involved in modulating the motor and sedative effects of cannabinoids. Preclinical and human genetic studies point towards a potential role of the ɑ5, ɑ3, and β4 nAChR subunits in CUD, while human GWAS studies strongly implicate the ɑ2 subunit as playing a role in CUD susceptibility. Clinical studies suggest that current smoking cessation agents, such as varenicline and bupropion, may also be beneficial in treating CUD, although more controlled studies are necessary. Additional behavioral, molecular, and mechanistic studies investigating the role of nAChR in the modulation of the pharmacological effects of cannabinoids are needed.
Pharmacological Research 191 (2023) 106746
Available online 29 March 2023
1043-6618/© 2023 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
Role of neuronal nicotinic acetylcholine receptors in
cannabinoid dependence
Belle Buzzi
a
,
*
, Eda Koseli
a
, Lauren Moncayo
a
, Mohammed Shoaib
b
, M Imad Damaj
a
a
Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
b
Newcastle, UK
ARTICLE INFO
Keywords:
Nicotine
Nicotinic acetylcholine receptors
Smoking
Cannabis
THC
Cannabis use disorder (CUD)
ABSTRACT
Cannabis is among the most widely consumed psychoactive drugs around the world and cannabis use disorder
(CUD) has no current approved pharmacological treatment. Nicotine and cannabis are commonly co-used which
suggests there to be overlapping neurobiological actions supported primarily by the co-distribution of both re-
ceptor systems in the brain. There appears to be strong rationale to explore the role that nicotinic receptors play
in cannabinoid dependence. Preclinical studies suggest that the ɑ7 nAChR subtype may play a role in modulating
the reinforcing and discriminative stimulus effects of cannabinoids, while the ɑ4β2 * nAChR subtype may be
involved in modulating the motor and sedative effects of cannabinoids. Preclinical and human genetic studies
point towards a potential role of the ɑ5, ɑ3, and β4 nAChR subunits in CUD, while human GWAS studies strongly
implicate the ɑ2 subunit as playing a role in CUD susceptibility. Clinical studies suggest that current smoking
cessation agents, such as varenicline and bupropion, may also be benecial in treating CUD, although more
controlled studies are necessary. Additional behavioral, molecular, and mechanistic studies investigating the role
of nAChR in the modulation of the pharmacological effects of cannabinoids are needed.
1. Introduction
Cannabis is among the most widely consumed psychoactive sub-
stances worldwide. An estimated 4% of the world population aged
1564 used cannabis at least once in 2019 (United Nations Ofce on
Drugs and Crime. [93].). In the United States, cannabis represents the
most abused federally illicit drug with 18% of the population reporting
past use (2020 NSDUH Detailed Tables | [12].). Studies estimate that
approximately 1030% of the 48 million Americans who use cannabis
are likely to develop cannabis use disorder (CUD) [34,47]. Those that
are diagnosed with CUD exhibit a continued problematic pattern of use
despite negative consequences, which causes signicant distress or
impairment in cognitive function [63]. CUD is also characterized by the
development of tolerance, and withdrawal upon cessation [34].
Δ9-Tetrahydrocannabinol (THC) is the main addictive component
in Cannabis sativa that acts in the central nervous system (CNS) by
binding to cannabinoid receptors (CB) in different brain areas to pro-
duce its ‘high, characterized by mild euphoria, relaxation, and
perceptual and cognitive alterations [29]. Additionally, clinical studies
have shown a similar array of withdrawal symptoms following absti-
nence from repeated oral THC ingestion or smoking cannabis [9,31].
Cannabis withdrawal during abstinence is comparative to that of to-
bacco or nicotine cessation, with reports of increased craving to resume
intake [18]. Despite the obvious societal implications of cannabis abuse,
mechanisms underlying THC dependence remain widely unknown, and
there are currently no FDA-approved medications for the treatment of
marijuana use disorder. With the dramatic shift in the legal, political,
and cultural landscape surrounding cannabis over the last decade, it is
likely that the number of individuals with CUD will increase in the
coming years as cannabis becomes more widely accepted and the
perceived risks continue to decline. Effective treatments for CUD are
clearly in need.
2. Neurobiology of cannabinoid dependence
The various pharmacological and behavioral effects of THC and
The multifaceted activities of nervous and non-nervous neuronal nicotinic acetylcholine receptors in physiology and pathology. Eds: Dr Cecilia Gotti, Prof
Francesco Clementi, Prof Michele Zoli
* Correspondence to: Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, PO BOX 980613, Richmond, VA
23298-0613, USA.
E-mail address: buzzib@vcu.edu (B. Buzzi).
Contents lists available at ScienceDirect
Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs
https://doi.org/10.1016/j.phrs.2023.106746
Received 21 December 2022; Received in revised form 15 March 2023; Accepted 27 March 2023
Pharmacological Research 191 (2023) 106746
2
cannabinoid agonists are mediated by the endocannabinoid system. The
endocannabinoid system is composed of two CB receptors (CB1 and
CB2) and two main endogenous ligands anandamide (AEA) and 2-arach-
idonoyl-glycerol (2-AG) and the associated enzymes responsible for
their synthesis and degradation. AEA and 2-AG are synthesized on de-
mand by their respective biosynthetic enzymes post-synaptically and
travel retrogradely to bind and activate pre-synaptic CB1 and CB2.
Additionally, endocannabinoid tone is highly regulated by several spe-
cic biosynthetic and metabolic enzymes. For example, AEA is synthe-
sized by N-acyl-phosphatidylethanolamine-specic phospholipase D
(NAPE-PLD) and later degraded by fatty acid amide hydrolase (FAAH).
Whereas diacylglycerol-lipases (DAGL
α
and DAGLβ) synthesize 2-AG
which is metabolized by monoacylglycerol-lipase (MAGL) [70].
2.1. Neurocircuitry of cannabinoid dependence
Both G
i
-coupled G-protein coupled receptors, CB1 and CB2 receptors
are understood to have specialized roles due to the high expression of
CB2 on peripheral immune cells and the primary expression of CB1 on
neurons in spinal cord and brain. Therefore, the effects of THC (among
other cannabinoids) and subsequent abuse potential are thought to be
mediated through central CB1 receptors. For example, activation of CB1
by ligands such as THC and its active metabolites activate the meso-
limbic reward pathway that regulates components of drug dependence
and withdrawal [70]. CB1 is highly expressed in cortical regions, with
the majority of mRNA levels detected in glutamatergic neurons [32,58].
Additionally, high levels of CB1 are also detected in GABAergic cells of
the striatum [43,51]. Therefore, CB1 is highly expressed in cells with
projections in dopaminergic pathways involved in drug abuse. In fact,
several CNS tissues and dopaminergic pathways are implicated in
cannabinoid tolerance, rewarding, and withdrawal-induced aversive--
like effects [26].
2.2. Interactions between nicotinic and cannabinoid systems in the brain
The endocannabinoid system shares anatomical and functional
similarities with nicotinic receptors. For example, rats tolerant to THC
show an increased endocannabinoid tone in limbic regions, such as the
amygdala, that express high levels of
α
7 nicotinic receptors [23,75].
Additionally,
α
7 expression overlaps in the cerebral cortex where
tolerance develops to THC metabolism following repeated administra-
tion [75,90]. Indeed, repeated activation of cannabinoid receptors by
agonists produces changes in brain regions with high expression of
several nicotinic receptor subtypes. Upon repeated activation, down-
regulation of CB1 occurs in the substantia nigra, a tissue with high
distribution of
α
5,
α
6 and β3 nicotinic receptor subunits. Although it is
clear the neurobiology of the endocannabinoid system interacts with the
nicotinic system in the brain, the exact relationship between nicotinic
receptor subtypes and cannabinoid dependence is not well organized.
3. Role of neuronal nicotinic receptors (nAChRs) in cannabinoid
dependence
A number of epidemiological and longitudinal studies showed that a
large number of cannabis users also identify as cigarette smokers, with
some suggesting that tobacco use may contribute to an increased inci-
dence of cannabis use disorder [4,67,69]. In addition, cannabis and
tobacco are noted as one of the most concurrently used drugs of abuse by
adolescents and young adults throughout the world [68]. The co-use of
tobacco and cannabis is frequent among cannabis users (4078% use a
tobacco product) and can occur simultaneously, sequentially or asyn-
chronously [61,73]. However, clinical and preclinical studies investi-
gating this co-use/abuse are challenging because of the diversity in
tobacco and cannabis use patterns and cannabis commercial products.
While the relationship between smoking and cannabis use is compli-
cated, it does suggest a possible interaction between the nicotinic and
endocannabinoid neuronal systems. Although several studies have
sought to determine the role of the endocannabinoid system in nicotine
dependence, the question on how the nicotinic cholinergic system plays
a role in cannabinoid dependence is not well studied [14,25,44,59]. This
review seeks to outline and examine the current preclinical and clinical
studies available on the role of neuronal nAChRs in cannabinoid
dependence, as well as discuss what is further needed to advance our
understanding of the interplay between both systems.
3.1. The nicotinic cholinergic system
The primary psychoactive ingredient with addictive properties in
tobacco is nicotine. Nicotine acts as an agonist for the pentameric
ligand-gated ion channel receptors known as nicotinic acetylcholine
receptors (nAChRs). Agonist binding to these receptors causes a
conformational change in the ion channel, allowing positively charged
ions to move across it and therefore a depolarization of the plasma
membrane. These nAChRs are distributed throughout the central and
peripheral nervous system; however, this review will focus on the
neuronal nAChR system [92]. Although nAChRs may contain a variety
of functional subtypes with differing compositions of ɑ (210) and β
subunits (24), the main nAChR subtypes responsible for nicotines
rewarding effects are ɑ4β2 * (the asterisk denotes the possible presence
of other subunits) as the primary heteromeric receptor subtype and ɑ7 as
the primary homomeric receptor subtype. nAChR kinetics and func-
tional properties vary in relation to subunit composition. In particular,
α
4β2 * nAChRs display a high afnity for ACh and nicotine and lower
Ca2 +-permeability. On the other hand,
α
7-nAChRs have a relatively
low afnity for ACh and nicotine and are highly Ca2 +-permeable [28].
Additionally, heteromeric receptor subtypes are thought to contain 2
ligand binding sites between ɑ and β subunits, while homomeric re-
ceptor subtypes contain 5 identical ligand binding sites [92].
A crosstalk between the nicotinic and cannabinoid systems is sub-
stantiated by the overlapping distribution of cannabinoid and nicotinic
acetylcholine receptors in many brain regions involved in drug reward
and dependence, such as the VTA, which project to the nucleus
accumbens (NAc), and to other limbic structures, such as the amygdala,
hippocampus, and prefrontal cortex. Activation of nAChRs within the
VTA stimulates the ring rate and bursting activity of dopamine neurons
and dopamine release, specically in the NAc, a common mechanism
underlying drug-induced reward and reinforcement [22,54,65]. In this
area, nAChRs are located not only on cell bodies of dopamine neurons
but also on GABAergic cell bodies and terminals and on presynaptic
nerve endings of glutamatergic neurons [13]. Studies have additionally
implicated the role of cannabinoids on altering glutamate transmission,
indicating another cross-over of both systems [16].
3.2. Preclinical studies
3.2.1. nAChR involvement in cannabinoid reward and reinforcement
Self-administration paradigms in preclinical models serve as a means
of interpreting the potential reinforcing effects of drugs of abuse [62].
Self-administration models in animals use operant conditioning in
which, typically, a lever press results in the delivery of a reinforcer, such
as a drug. Drugs may be delivered using intravenous, oral, inhalation, or
insufation routes, among others. Intravenous (i.v.) self-administration
of Δ9-THC has been developed in non-human primates [40,83], and in
rodents [81,82]. I.v. self-administration was reported for other CB1 re-
ceptor agonists as well, such as WIN55,2122, in rodents [3,41,80,89].
Recently, nicotine injections were reported to increase i.v.
self-administration of WIN55,2122 and THC in adult rats, implicating a
potential role of nAChRs in cannabinoid self-administration [82]. As
both nAChRs and CB1Rs are colocalized in several brain areas involved
in reward, questions remain about the crosstalk mechanisms of both
systems in a model of cannabinoid self-administration. However, the
nAChR subtypes mediating this interaction is unknown.
B. Buzzi et al.
Pharmacological Research 191 (2023) 106746
3
Both ɑ4β2 * and ɑ7 nAChRs are highly expressed in regions of the
brain involved in reward, such as the mesolimbic dopaminergic system,
however Solinas and others suggest that the homomeric ɑ7 nAChRs, and
not ɑ4β2 * nAChRs, modulate the abuse-related behaviors and neuro-
chemical changes of cannabinoid dependence [79]. Using a relatively
selective ɑ7 antagonist, methyllycaconitine (MLA), they found that
systemic injections of MLA signicantly decreased intravenous
self-administration of CB1 agonist WIN55,2122 in rats, while dihy-
dro-β-erythroidine (DHβE), a nAChR selective β2 * antagonist, had no
effect on WIN55,2122 self-administration. Furthermore, MLA was able
to block THC-induced increases in dopamine levels in the nucleus
accumbens (NAc) shell, while DHβE did not. Similarly, another study
found that neuronal increase of kynurenic acid (KYNA), a non-selective
ɑ7 nAChR negative allosteric modulator (NAM), by a selective kynur-
enine 3-monooxygenase (KMO) inhibitor Ro 618048, signicantly
decreased i.v. self-administration of WIN55,2122 and THC in rats and
squirrel monkeys, respectively [42]. Ro 618048 additionally prevented
a relapse-like effect induced by a priming dose of WIN55,2122 in rats.
Neurochemically, Ro 618048 was able to block THC and WIN55,
2122-induced elevations of dopamine in the NAc shell in rats. Pre-
treatment with ɑ7 nAChR positive allosteric modulators galantamine
and PNU120596 was able to reverse all of these behavioral and neuro-
chemical blockades by Ro 618048, further suggesting a role of ɑ7
nAChRs in cannabinoid intake and relapse in animal models [42].
Conditioned place preference (CPP) is another behavioral model
used to assess the potential rewarding effects of drugs using classical
conditioning learning paradigm [52]. In preclinical research, the basic
premise involves pairing the drug of interest, the unconditioned stim-
ulus, with an initially neutral environment, the conditioned stimulus,
typically either a dark side or a light side of a box, with differing tactile
stimuli, over several conditioning days. The subjectspreference is then
tested in a drug-free state where they are allowed to freely explore both
sides over a period of time, typically measuring preference as the time
spent on the drug-paired side compared to either the vehicle-paired side
or the baseline preference. One study explored the effect of seven-week
exposure to nicotine, using either electronic cigarette (e-cig) delivery or
cigarette (cig) delivery, on THC-induced expression of preference using
the CPP paradigm in Balb/c mice [66]. They found that pre-exposure to
nicotine, using both e-cig and cig delivery increased sensitivity to THCs
rewarding effects using a sub-threshold dose of THC that did not cause
an expression of preference in both naive mice and those pre-exposed to
air for seven weeks [66]. Additionally, they observed an increase in
ΔFosB in the NAc, as well as an increase in the AMPA receptor
GluA1/GluA23 subunit ratio in the groups that received pre-exposure
to nicotine, hypothesizing that these changes may be responsible for
the increased sensitivity to THCs rewarding effects [66]. However, the
nAChR subtypes mediating the effects of nicotine are not known.
3.2.2. nAChR involvement in cannabinoid drug discrimination
Drug discrimination in animals is a preclinical paradigm that uses
operant conditioning to model the subjective, pharmacological, and
discriminative properties of drugs [94]. In short, the model uses drugs as
discriminative stimuli in order to signal the delivery of a reinforcer and
measures the ability of the animal to identify the drug of choice when
compared to a vehicle or other drugs [30].
The studies that examined the role of ɑ7 nAChRs versus non-ɑ7
nAChRs in cannabinoid self-administration also evaluated the ability of
ɑ7 and non-ɑ7 allosteric modulators to alter the discriminative prop-
erties of cannabinoids. In rats that were able to discriminate the effects
of both THC and WIN55,2122, the ɑ7 antagonist MLA dose-
dependently blocked the discriminative stimulus of THC and
WIN55,2122, while DHβE was not effective [79]. Ro 618048, a KMO
inhibitor that results in an increase in the ɑ7 negative allosteric modu-
lator KYNA, was conversely not able to block the discriminative effects
of WIN55,2122 in rats but was able to cause a rightward shift of the
dose-response curve of the discriminative effects of THC in squirrel
monkeys, indicating a possible decrease in potency following Ro
618048 administration [42]. This study demonstrates a potential role
in the ɑ7 nAChR in modulating the subjective effects of CB1R agonists in
rats and squirrel monkeys, however additional studies are needed to
further conrm this.
In a separate study, researchers sought to determine whether both
the nAChR and muscarinic cholinergic systems (mAChR) are involved in
the modulation of cannabinoid discrimination in rats and to elucidate
the potential mechanism [79]. Both the nAChR agonist nicotine and the
mAChR agonist pilocarpine were able to potentiate the discriminative
stimulus effects of low doses of THC, without decreasing rates of
responding. After determining that nAChRs play a role in the modula-
tion of THC discrimination, they used the CB1 antagonist rimonabant to
reverse this potentiation, which was only successful in nicotine, and not
pilocarpine-induced potentiation. The ability of the CB1 antagonist to
block nicotine-induced potentiation of THC discrimination suggests that
this potentiation by nicotine may be due to release of endocannabinoids.
To further delineate this theory, a selective fatty acid amide hydrolase
(FAAH) inhibitor, URB-597, which increases levels of the endocanna-
binoid anandamide, signicantly increased nicotine-induced potentia-
tion of THC discrimination, while URB-597 had no effect on
pilocarpine-induced potentiation of THC discrimination, conrming
the idea that nicotinic receptor activation potentiates THC discrimina-
tion via endocannabinoid release [79].
3.2.3. nAChR involvement in cannabinoid withdrawal
Chronic cannabinoid use followed by discontinuation has been re-
ported to cause cannabinoid withdrawal syndrome in both humans and
animals, a major factor behind users not being able to quit successfully
[9,26]. There are currently no approved medications to treat cannabis
use disorder or cannabis withdrawal. Early investigations on THC and
nicotinic system interaction showed that repeated co-administration of
nicotine and THC led to an attenuation in tolerance to THC but with
enhancement of somatic signs of precipitated THC withdrawal in CD-1
mice [85]. As nicotine and cannabinoids are often concurrently used,
the question remains on how the nicotinic system may play a role in
cannabinoid withdrawal, including the specic receptor subtypes
involved.
Nicotinic receptors containing
α
3,
α
5 and β4 subunits were investi-
gated for their role in cannabis withdrawal. Human genetic studies
showed that genetic variation in the
α
5/
α
3/β4 nAChR subunit gene
cluster signicantly increases the risk of tobacco addiction. In particular,
polymorphisms in the
α
5 subunit gene (CHRNA5) present compelling
evidence for a genetic contribution to tobacco addiction vulnerability
and smoking-related diseases ([5]; for review see [6]). In addition, ro-
dent studies implicated these subunits in nicotine withdrawal [37,36].
Given these ndings, our recent study investigated the role of these
subunits in THC withdrawal [24]. We found that a selective ɑ3β4
antagonist, AulB, and a partial ɑ3β4 agonist, AT-1001, both
dose-dependently attenuated precipitated THC withdrawal somatic
signs in C57BL/6 J mice. We also observed that ɑ5 KO mice showed a
decrease in precipitated THC withdrawal somatic signs, implicating ɑ5,
ɑ3, and β4 nAChR subunits in THC withdrawal. Additionally, other
studies showed that the ɑ6 nAChR subunit, expressed in dopaminergic
neurons which co-assembles with β4 and β2 containing receptor sub-
types [17,45] may play a role in THC withdrawal. ɑ6 KO mice and mice
given BulA, an ɑ6β4 antagonist, showed reduced precipitated THC
withdrawal somatic signs, implicating the ɑ6 nAChR subunit in THC
withdrawal. However, ɑ7 and β2 KO mice showed no differences in the
intensity of THC of somatic signs of withdrawal. While these studies
indicate an important role of the ɑ5, ɑ3, ɑ6, and β4 nAChR subunits in
THC withdrawal, clear limitations exist in the rodent model of precipi-
tated withdrawal in contrast to spontaneous cessation in humans.
Therefore, the extent to which these symptoms offer translational rele-
vance is unclear. Likewise, THC plasma levels were not reported in these
studies, so it is unknown if these doses used for THC administration are
B. Buzzi et al.
Pharmacological Research 191 (2023) 106746
4
correlated to human THC intake.
3.2.4. nAChR involvement in adverse behavioral effects of cannabinoids
Cannabis use comes with several side effects, some of which may be
undesirable. Side effects are comprised of behavioral and physical
manifestations. As an example, cannabinoids are known to cause a
disruption in working memory, attention impairments, increased anxi-
ety, insomnia, etc. [10,15]. One theory as to why cannabinoid users tend
to also use nicotine, and vice versa, is that they may both decrease these
adverse effects and increase the reinforcing effects of one another [25,
85]. A study found that chronic nicotine pretreatment reduced the
anxiogenic-like effects that are induced by high THC doses in both the
elevated plus maze (EPM) test and social interaction (SI) test in rats,
with the study suggesting that chronic nicotine may induce neuroplastic
alterations that modulate the response to THC exposure [50].
In addition to increased anxiety-like behavior, repeated cannabinoid
use is speculated to cause working memory impairments in both pre-
clinical and clinical models [20,48]. One study found that THC
decreased working memory in a non-matching-to-position model in rats
and that administration of ɑ7 NAM had no effect on this, while in
squirrel monkeys, a THC-induced decrease in working memory using a
delayed matching-to-sample model was reversed by the same ɑ7 NAM,
suggesting a possible role of the ɑ7 nAChR in the modulation of
THC-induced changes in working memory [42].
Cannabinoid use also causes a myriad of motor impairment effects,
including hypolocomotion, catalepsy, and ataxia, the latter of which can
be undesirable, especially when considering cannabinoids for treatment
and/or therapy of motor disorders [55]. Interestingly, chronic nicotine
exposure attenuated acute THC-induced hypolocomotion in adult rats,
raising the possibility that chronic nicotine sensitizes neurons in the
mesolimbic dopaminergic system, thus altering the responses to THC on
this system [57]. Additionally, it is known that repeated nicotine
exposure causes an increase in endocannabinoids in the VTA and NAc,
which may also play a role in this modulation [27].
In addition, acute THC administration is known to cause cerebellar
ataxia, among other motor effects, largely due to a population of CB1
receptors in the cerebellum [71,77]. The impact of nicotine on canna-
binoids motor suppressant effects was investigated early on since there
is a population of nAChRs expressed in the cerebellum as well [19].
Indeed, intracerebellar micro infusion of THC in CD-1 male mice caused
ataxia, which was dose-dependently attenuated by micro infusions of
nicotine [77]. Central administration of hexamethonium, a
non-selective peripheral nAChR antagonist, was able to block this
attenuation, demonstrating a role of the nicotinic receptors in this
modulation of THC-induced ataxia. RJR-2403, a selective ɑ4β2 * nAChR
agonist, was also able to dose-dependently attenuate this ataxia, which
was then blocked by the selective β2 * nAChR antagonist DhβE, impli-
cating specically that receptor subtype in this attenuation of
THC-induced ataxia. MLA, a selective ɑ7 nAChR antagonist, was also
tested against both nicotine and RJR, ultimately having no effect on the
attenuation, showing that ɑ7 plays no role in nicotine attenuation of
THC-induced ataxia [77].
After conrmation that the ɑ4β2 nAChR plays a direct role in nico-
tines attenuation of THC-induced ataxia, the mechanism of this atten-
uation was further explored by the same team [78]. nAChR activation
has been implicated in long-term potential (LTP) via the nitric
oxide-cGMP signaling pathway, and there are high nitric oxide synthase
(NOS) levels in the cerebellum [33]. As nicotine-induced modulation of
locomotor activity has been previously shown to be blocked by inhibi-
tion of nitric oxide synthase (NOS) and CB1 agonists have been shown to
inhibit NOS in the cerebellum [76], the same group sought to explore the
role of this system in THC cerebellar ataxia [78]. Using intracerebellar
infusions of sodium nitroprusside (SNP), a nitric oxide (NO) donor,
THC-induced ataxia was attenuated, and RJR attenuation of
THC-induced ataxia was even further enhanced when infused with SNP,
revealing a possible additive effect, or potential shared mechanism, of
increased NO and ɑ4β2 receptor activation on attenuation of
THC-induced ataxia. Furthermore, S-methylisothiourea (SMT), a NOS
inhibitor, inhibited RJR-induced attenuation of THC-induced ataxia as
well as increased THC-induced ataxia when given alone. As NO donors
act on guanylyl cyclase (GC) to increase muscle relaxation, the re-
searchers then looked at increasing levels of GC activation on nicotine
attenuation of THC-induced ataxia. Intracerebellar infusions of iso-
liquiritigenin (ISO), a GC activator, and ODQ, a GC inhibitor, respec-
tively enhanced and antagonized RJR attenuation of THC-induced
ataxia [78]. Additionally, NOx levels were conrmed to be lowered after
acute THC administration in mouse cerebellar tissue using the uoro-
metric DAN method, and increased after both nicotine and RJR
administration, showing a clear role of NO donors and GC activation on
RJR attenuation of THC- induced ataxia [78]. Overall, nicotine seems to
play a role via ɑ4β* 2 nAChR subtypes through increased NO and GC
activation, to attenuate the ataxia caused by acute administration of
THC. All preclinical studies are summarized in Table 1A.
3.3. Human studies
3.3.1. Clinical studies
Many tobacco users also misuse cannabis, and the concurrent use of
these two substances can add to the health burden experienced by such
cannabis users and may increase the potential for substance-induced
harm and disorders above that caused by using one substance on its
own. Therefore, recent studies explored the possible effectiveness of
medications used in smoking cessation, such as varenicline and bupro-
pion, for the treatment of cannabis use disorder (CUD). Varenicline, an
α
4β2 subtype partial agonist and a full agonist at the
α
3β4 and
α
7 sub-
types [56], is the current most effective approved pharmacotherapy for
tobacco smoking cessation (see [39] for review).
Initially, a case series reported reductions in amount of enjoyment of
cannabis and self-reported cannabis use among ve cannabis- and
nicotine-dependent individuals receiving varenicline for 12 weeks [60].
In a later laboratory study, Herrmann et al. [35] demonstrated that
varenicline reduced tobacco use, craving, and negative affect in tobac-
co/cannabis co-users, although it had no effect on cannabis relapse. In
addition, a small pilot trial reported reduced cannabis craving, cannabis
use, and tobacco use when varenicline was given to individuals with
opioid use disorder for four weeks [1]. Finally, a recent proof-of-concept
pilot randomized trial assessed the initial efcacy of varenicline
(6-weeks trial) in cannabis-using individuals and found signicant de-
creases in self-reported cannabis withdrawal and greater rates of
self-reported abstinence [53].
A small number of studies looked at the efcacy of bupropion, an
antidepressant, and a smoking cessation agent [91] for the treatment of
cannabis dependence. In a double-blind and placebo-controlled study
looking at bupropion-sustained release in a 13-week outpatient treat-
ment program, there was no evidence for a signicant increase in the
probability of abstinence from cannabis or a reduction of the severity of
cannabis dependence and cannabis withdrawal symptoms [11]. How-
ever, bupropion was found to be effective in reducing symptoms expe-
rienced in marijuana withdrawal in a double-blind, placebo-controlled
study with chronic marijuana users [64]. All clinical studies are sum-
marized in Table 1B.
3.3.2. Human genetic studies
Based on several twin and family heritability studies, the heritability
of CUD initiation is estimated to be 3070% which vary largely due to
environment factors [49,87,88]. Recent genome-wide association
studies (GWASs) of CUD have identied variants reaching genome-wide
signicance, with one locus on chromosome 8, tagged by a cis-eQTL for
CHRNA2 (encoding a nicotinic acetylcholine receptor), has been
robustly identied [21]. In addition, they found that individuals with
CUD show reduced expression of CHRNA2 in the cerebellum compared
to the controls. The locus on chromosome 8 was recently conrmed in a
B. Buzzi et al.
Pharmacological Research 191 (2023) 106746
5
large GWAS meta-analysis [38]. The comparison of the expression
proles in the Allan Brain Atlas [7], shows a negative correlation on the
cerebellar cortex and cerebellar nuclei between CHRNA2 and CNR1
(cannabinoid receptor 1) gene expressions. These ndings suggest that
CHRNA2 expression which encodes for the
α
2 nAChR subunit may play
a functional role in susceptibility to developing CUD. Interestingly
CHRNA2 gene has been identied as one of the risk loci for nicotine
dependence and smoking behavior [46] and
α
2 nAChR KO mice show
reduced precipitated nicotine withdrawal signs [72]. Finally, in an as-
sociation study using the Gene and Environment (SAGE)
European-American and African-American datasets, our group investi-
gated the correlation between cannabis dependence and nAChRs [24].
The results of the study suggested that the following genes that code
nAChRs subunits were associated with CUD;
α
3 (CHRNA3) with toler-
ance,
α
5 (CHRNA5) with withdrawal, and
α
6 (CHRNA6) with both
tolerance and withdrawal. In addition, the correlation between CHRNA3
and CHRNA5 genes and CUD was signicant using the DSM-IV criteria
[24].
4. Looking ahead
Cannabis remains the most abused federally illegal drug in the
United States and is heavily used around the world. CUD, which is
characterized by continuous use of cannabis (or other cannabinoids)
despite negative consequences, has yet to have an approved pharma-
cological treatment. As nicotine is often co-used with cannabis and with
the overlapping distribution of CBRs and nAChRs in brain regions
involved in substance abuse, the role that the nicotinic system plays in
cannabis use and abuse represents a high area of research interest.
Preclinical studies suggest that the ɑ7 nAChR subtype may play a
role in modulating both the reinforcing and discriminative stimulus ef-
fects of cannabinoids, and in the modulation of THC-induced decrease in
working memory, using both behavioral and neurochemical assays.
Further studies exploring the role of this nAChR subtype in cannabinoid
drugs seeking, reinstatement, and choice preferences are necessary. For
that, developing selective ɑ7 NAMs with drug-like properties for clinical
use is needed. While some preclinical studies listed here used orthosteric
ɑ7 antagonists, this use may be problematic in clinical applications as
early rodent studies suggest that these drugs may cause working mem-
ory impairments [2]. Alternatively, ɑ7 partial agonists could be tested in
preclinical cannabis animal models for their potential efcacy. Several
ɑ7 partial agonists that were tested in clinical studies for schizophrenia.
Such as Encenicline and ABT-126 are possible candidates [84]. KMO
inhibitors, which increase levels of the ɑ7 NAM KYNA, is currently being
investigated as a treatment for other substances of abuse in preclinical
models [74,86], and for schizophrenia in clinical trials [8]. However, it
will be important to be further explored in its role in CUD. While the ɑ7
nAChR has been implicated in the rewarding effects of cannabinoids, via
endocannabinoid release, the ɑ4β2 * nAChR has been implicated in the
attenuation of cannabinoid induced motor impairments, via increasing
NO levels, suggesting that there may be differential roles of the neuronal
nicotinic acetylcholine system in the behavioral and neurochemical
modulation of the effects of cannabinoids.
Both preclinical and human genetic studies indicate a potential role
of ɑ5, ɑ3, and β4 nAChR subunits in CUD phenotypes, including toler-
ance and withdrawal. Human GWAS studies strongly implicated the ɑ2
subunit as playing a possible functional role in both susceptibility to
CUD and in nicotine withdrawal. Clearly the role of CHRNA2 gene needs
to be elucidated further in terms of its role in cannabinoid pharmacology
and dependence. As there is no current selective ligand for the ɑ2
subunit-containing subtypes, there is a need to determine not only which
set of subunits play a role in cannabinoid dependence but also the
location of these subtypes that are thought to be correlated with
increased susceptibility of CUD. Additionally, as both ɑ3β4 and ɑ5
containing subunits were implicated as playing a role THC withdrawal
in preclinical and genetic studies, these should also be considered a
potential target to further explore in CUD.
Smoking cessation agents that target nicotinic receptors, such as
Table 1A
Preclinical studies.
Behavioral Test Species Strain Sex Drugs Exposure Result Reference
THC Elevated Plus Maze Rats Sprague-Dawley M/
F
Nicotine 14 days [50]
THC Social Interaction Rats Sprague-Dawley M/
F
Nicotine 14 days [50]
Somatic Signs of Precipitated
THC Withdrawal
Mice C57BL/6 J M ɑ3β4 antagonist AulB, ɑ3β4 partial agonist AT-1001, ɑ6β4
antagonist BulA
Single
dose
[24]
Self-administration of THC
and WIN55,2122
Rats, Squirrel
Monkeys
Sprague-Dawley,
Long-Evans,
M KMO inhibitor Ro 618048, β2 * antagonist DHβE, ɑ7
antagonist MLA, Nicotine, ɑ7 PAMs galantamine and
PNU120596
Mixed / [42,79,
82]
Drug Discrimination of THC
and WIN55,2122
Rats Sprague-Dawley M Nicotine, Pilocarpine, ɑ7 antagonist MLA, β2 * antagonist
DHβE
Single
dose
/ [79]
Rotorod (THC Induced
Ataxia)
Mice CD-1 M Nicotine, ɑ3β4 agonist RJR-2403, Single
dose
[77]
THC CPP Mice Balb/c M Nicotine 7 weeks [66]
THC Locomotor Activity Rats Sprague-Dawley M/
F
Nicotine 14 days [57]
=reduced response; =increased response; =no reliable response, /=mixed responses
Table 1B
Clinical studies.
Outcomes Drug of Abuse Sex Drug Tested Exposure Result Reference
Usage Nicotine and/or Cannabis M Varenicline (Chantix®) 26 weeks [60]
Mood, Cravings, Sleep, Usage Tobacco and Cannabis Co-users M/F Varenicline, Nabilone 16 days / [35]
Usage, Cravings, Withdrawal Co-occurring Cannabis and Tobacco use M/F Varenicline 4 weeks [1]
Usage, Withdrawal Cannabis M/F Varenicline 6 weeks [53]
Craving, Withdrawal, Sleep, Cognition Cannabis M/F Bupropion
(Zyban® SR)
21 days / [64]
Usage and Withdrawal Cannabis M/F Bupropion, Nefazodone 13 weeks [11]
=reduced response; =increased response; =no reliable response, /=mixed responses
B. Buzzi et al.
Pharmacological Research 191 (2023) 106746
6
varenicline and bupropion, have been investigated in the treatment of
CUD and while the pilot studies showed promising results, large and
controlled clinical trials are essential. Overall, further behavioral studies
and molecular mechanisms of the role that nAChRs play in the modu-
lation of the rewarding and aversive effects of cannabinoids are needed.
CRediT authorship contribution statement
B. Buzzi: Conceptualization, Visualization, Writing - original draft,
Writing - review & editing. E. Koseli: Visualization, Writing - original
draft, Writing - review & editing. L. Moncayo: Writing - original draft,
Writing - review & editing. M. Shoaib: Writing - original draft, Writing -
review & editing. M.I. Damaj: Conceptualization, Supervision, Funding
acquisition, Writing - original draft, Writing - review & editing.
Declarations of Competing Interest
No conict of interests.
Data Availability
No data was used for the research described in the article.
References
[1] T.R. Adams, J.H. Arnsten, Y. Ning, S. Nahvi, Feasibility and preliminary
effectiveness of varenicline for treating co-occurring cannabis and tobacco use,
J. Psychoact. Drugs 50 (1) (2018) 1218, https://doi.org/10.1080/
02791072.2017.1370746.
[2] N.A. Addy, A. Nakijama, E.D. Levin, Nicotinic mechanisms of memory: effects of
acute local DHbetaE and MLA infusions in the basolateral amygdala. Brain
Research, Cogn. Brain Res. 16 (1) (2003) 5157, https://doi.org/10.1016/s0926-
6410(02)00209-4.
[3] P. Amchova, J. Kucerova, V. Giugliano, Z. Babinska, M. Zanda, M. Scherma,
L. Dusek, P. Fadda, V. Micale, A. Sulcova, W. Fratta, L. Fattore, Enhanced self-
administration of the CB1 receptor agonist WIN55,212-2 in olfactory
bulbectomized rats: evaluation of possible serotonergic and dopaminergic
underlying mechanisms, Front. Pharmacol. 5 (2014). https://www.frontiersin.or
g/articles/10.3389/fphar.2014.00044.
[4] A. Amos, S. Wiltshire, Y. Bostock, S. Haw, A. McNeill, You cant go without a fag.
you need it for your hash’—a qualitative exploration of smoking, cannabis and
young people, Addiction 99 (1) (2004) 7781, https://doi.org/10.1111/j.1360-
0443.2004.00531.x.
[5] L.J. Bierut, Nicotine dependence and genetic variation in the nicotinic receptors,
Drug Alcohol Depend. 104 (Suppl 1) (2009) S64S69, https://doi.org/10.1016/j.
drugalcdep.2009.06.003.
[6] L.J. Bierut, Convergence of genetic ndings for nicotine dependence and smoking
related diseases with chromosome 15q24-25, Trends Pharmacol. Sci. 31 (1) (2010)
4651, https://doi.org/10.1016/j.tips.2009.10.004.
[7] Brain Map Brain-map.org. (n.d.). Retrieved March 15, 2023, from https://
portal.brain-map.org/.
[8] Buchanan, R. (2022). The Effects of Kynurenine Aminotransferase Inhibition in
People With Schizophrenia (Clinical Trial Registration No. NCT04013555).
clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT04013555.
[9] A.J. Budney, J.R. Hughes, The cannabis withdrawal syndrome, Curr. Opin.
Psychiatry 19 (3) (2006) 233238, https://doi.org/10.1097/01.
yco.0000218592.00689.e5.
[10] E.J. Calabrese, A. Rubio-Casillas, Biphasic effects of THC in memory and cognition,
Eur. J. Clin. Investig. 48 (5) (2018), e12920, https://doi.org/10.1111/eci.12920.
[11] K.M. Carpenter, D. McDowell, D.J. Brooks, W.Y. Cheng, F.R. Levin, A preliminary
trial: double-blind comparison of nefazodone, bupropion-SR, and placebo in the
treatment of cannabis dependence, Am. J. Addict. 18 (1) (2009) 5364, https://
doi.org/10.1080/10550490802408936.
[12] CBHSQ Data. (n.d.). Retrieved June 16, 2022, from https://www.samhsa.gov/
data/report/ 2020 -nsduh-detailed-tables.
[13] J.-P. Changeux, Nicotine addiction and nicotinic receptors: lessons from genetically
modied mice, Nat. Rev. Neurosci. 11 (6) (2010) 389401, https://doi.org/
10.1038/nrn2849.
[14] X. Chen, V.S. Williamson, S.-S. An, J.M. Hettema, S.H. Aggen, M.C. Neale, K.
S. Kendler, Cannabinoid receptor 1 gene association with nicotine dependence,
Arch. Gen. Psychiatry 65 (7) (2008) 816824, https://doi.org/10.1001/
archpsyc.65.7.816.
[15] K. Cohen, A. Weinstein, The effects of cannabinoids on executive functions:
evidence from cannabis and synthetic cannabinoidsa systematic review, Brain
Sci. 8 (3) (2018) 40, https://doi.org/10.3390/brainsci8030040.
[16] M. Colizzi, P. McGuire, R.G. Pertwee, S. Bhattacharyya, Effect of cannabis on
glutamate signalling in the brain: a systematic review of human and animal
evidence, Neurosci. Biobehav. Rev. 64 (2016) 359381, https://doi.org/10.1016/j.
neubiorev.2016.03.010.
[17] G. Collo, L. Cavalleri, M. Zoli, U. Maskos, E. Ratti, E. Merlo Pich, Alpha6-containing
nicotinic acetylcholine receptors mediate nicotine-induced structural plasticity in
mouse and human iPSC-derived dopaminergic neurons, Front. Pharmacol. 9 (2018)
572, https://doi.org/10.3389/fphar.2018.00572.
[18] J.R. Cornelius, T. Chung, C. Martin, D.S. Wood, D.B. Clark, Cannabis withdrawal is
common among treatment-seeking adolescents with cannabis dependence and
major depression, and is associated with rapid relapse to dependence, Addict.
Behav. 33 (11) (2008) 15001505, https://doi.org/10.1016/j.
addbeh.2008.02.001.
[19] G. De Filippi, T. Baldwinson, E. Sher, Evidence for nicotinic acetylcholine receptor
activation in rat cerebellar slices, Pharmacol. Biochem. Behav. 70 (4) (2001)
447455, https://doi.org/10.1016/S0091-3057(01)00653-0.
[20] L. Dellazizzo, S. Potvin, S. Gigu`
ere, A. Dumais, Evidence on the acute and residual
neurocognitive effects of cannabis use in adolescents and adults: a systematic meta-
review of meta-analyses, Addiction 117 (7) (2022) 18571870, https://doi.org/
10.1111/add.15764.
[21] D. Demontis, V.M. Rajagopal, T.E. Thorgeirsson, T.D. Als, J. Grove, K. Lepp¨
al¨
a, D.
F. Gudbjartsson, J. Pallesen, C. Hjorthøj, G.W. Reginsson, T. Tyrngsson,
V. Runarsdottir, P. Qvist, J.H. Christensen, J. Bybjerg-Grauholm, M. Bækvad-
Hansen, L.M. Huckins, E.A. Stahl, A. Timmermann, A.D. Børglum, Genome-wide
association study implicates CHRNA2 in cannabis use disorder, Nat. Neurosci. 22
(7) (2019) 10661074, https://doi.org/10.1038/s41593-019-0416-1.
[22] G. Di Chiara, A. Imperato, Drugs abused by humans preferentially increase synaptic
dopamine concentrations in the mesolimbic system of freely moving rats, Proc.
Natl. Acad. Sci. 85 (14) (1988) 52745278, https://doi.org/10.1073/
pnas.85.14.5274.
[23] V. Di Marzo, F. Berrendero, T. Bisogno, S. Gonz´
alez, P. Cavaliere, J. Romero,
M. Cebeira, J.A. Ramos, J.J. Fern´
andez-Ruiz, Enhancement of anandamide
formation in the limbic forebrain and reduction of endocannabinoid contents in the
striatum of delta9-tetrahydrocannabinol-tolerant rats, J. Neurochem. 74 (4) (2000)
16271635, https://doi.org/10.1046/j.1471-4159.2000.0741627.x.
[24] G. Donvito, P.P. Muldoon, K.J. Jackson, U. Ahmad, N.T. Zaveri, J.M. McIntosh,
X. Chen, A.H. Lichtman, M.I. Damaj, Neuronal nicotinic acetylcholine receptors
mediate Δ9-THC dependence: mouse and human studies, Addict. Biol. 25 (1)
(2020), e12691, https://doi.org/10.1111/adb.12691.
[25] I. Gamaleddin, C. Wertheim, A.Z.X. Zhu, K.M. Coen, K. Vemuri, A. Makryannis, S.
R. Goldberg, B. Le Foll, Cannabinoid receptor stimulation increases motivation for
nicotine and nicotine seeking, Addict. Biol. 17 (1) (2012) 4761, https://doi.org/
10.1111/j.1369-1600.2011.00314.x.
[26] S. Gonz´
alez, M. Cebeira, J. Fern´
andez-Ruiz, Cannabinoid tolerance and
dependence: a review of studies in laboratory animals, Pharmacol., Biochem.,
Behav. 81 (2) (2005) 300318, https://doi.org/10.1016/j.pbb.2005.01.028.
[27] S. Gonz´
alez, M.G. Cascio, J. Fern´
andez-Ruiz, F. Fezza, V. Di Marzo, J.A. Ramos,
Changes in endocannabinoid contents in the brain of rats chronically exposed to
nicotine, ethanol or cocaine, Brain Res. 954 (1) (2002) 7381, https://doi.org/
10.1016/s0006-8993(02)03344-9.
[28] C. Gotti, F. Clementi, A. Fornari, A. Gaimarri, S. Guiducci, I. Manfredi, M. Moretti,
P. Pedrazzi, L. Pucci, M. Zoli, Structural and functional diversity of native brain
neuronal nicotinic receptors, Biochem. Pharmacol. 78 (7) (2009) 703711, https://
doi.org/10.1016/j.bcp.2009.05.024.
[29] B. Green, D. Kavanagh, R. Young, Being stoned: a review of self-reported cannabis
effects, Drug Alcohol Rev. 22 (4) (2003) 453460, https://doi.org/10.1080/
09595230310001613976.
[30] J. H¨
aggkvist, J. Franck, Chapter 7 - animal models of addiction other than alcohol:
amphetamines, in: P.M. Miller (Ed.), Biological Research on Addiction, Academic
Press, 2013, pp. 6168, https://doi.org/10.1016/B978-0-12-398335-0.00007-8.
[31] M. Haney, A.S. Ward, S.D. Comer, R.W. Foltin, M.W. Fischman, Abstinence
symptoms following smoked marijuana in humans, Psychopharmacology 141 (4)
(1999) 395404, https://doi.org/10.1007/s002130050849.
[32] T. Harkany, M.B. Dobszay, F. Cayetanot, W. H¨
artig, T. Siegemund, F. Aujard,
K. Mackie, Redistribution of CB1 cannabinoid receptors during evolution of
cholinergic basal forebrain territories and their cortical projection areas: a
comparison between the gray mouse lemur (Microcebus murinus, primates) and
rat, Neuroscience 135 (2) (2005) 595609, https://doi.org/10.1016/j.
neuroscience.2005.06.043.
[33] N.A. Hartell, Receptors, second messengers and protein kinases required for
heterosynaptic cerebellar long-term depression, Neuropharmacology 40 (1) (2001)
148161, https://doi.org/10.1016/S0028-3908(00)00107-6.
[34] D.S. Hasin, T.D. Saha, B.T. Kerridge, R.B. Goldstein, S.P. Chou, H. Zhang, J. Jung,
R.P. Pickering, W.J. Ruan, S.M. Smith, B. Huang, B.F. Grant, Prevalence of
marijuana use disorders in the united states between 2001-2002 and 2012-2013,
JAMA Psychiatry 72 (12) (2015) 12351242, https://doi.org/10.1001/
jamapsychiatry.2015.1858.
[35] E.S. Herrmann, Z.D. Cooper, G. Bedi, D. Ramesh, S.C. Reed, S.D. Comer, R.
W. Foltin, M. Haney, Varenicline and nabilone in tobacco and cannabis co-users:
effects on tobacco abstinence, withdrawal and a laboratory model of cannabis
relapse, Addict. Biol. 24 (4) (2019) 765776, https://doi.org/10.1111/adb.12664.
[36] K.J. Jackson, S.S. Sanjakdar, P.P. Muldoon, J.M. McIntosh, M.I. Damaj, The
α
3β4*
nicotinic acetylcholine receptor subtype mediates nicotine reward and physical
nicotine withdrawal signs independently of the
α
5 subunit in the mouse,
Neuropharmacology 70 (2013) 228235, https://doi.org/10.1016/j.
neuropharm.2013.01.017.
[37] K.J. Jackson, M.J. Marks, R.E. Vann, X. Chen, T.F. Gamage, J.A. Warner, M.
I. Damaj, Role of
α
5 nicotinic acetylcholine receptors in pharmacological and
B. Buzzi et al.
Pharmacological Research 191 (2023) 106746
7
behavioral effects of nicotine in mice, J. Pharmacol. Exp. Ther. 334 (1) (2010)
137146, https://doi.org/10.1124/jpet.110.165738.
[38] E.C. Johnson, D. Demontis, T.E. Thorgeirsson, R.K. Walters, R. Polimanti, A.
S. Hatoum, S. Sanchez-Roige, S.E. Paul, F.R. Wendt, T.-K. Clarke, D. Lai, G.
W. Reginsson, H. Zhou, J. He, D.A.A. Baranger, D.F. Gudbjartsson, R. Wedow, D.
E. Adkins, A.E. Adkins, A. Agrawal, A large-scale genome-wide association study
meta-analysis of cannabis use disorder, Lancet Psychiatry 7 (12) (2020)
10321045, https://doi.org/10.1016/S2215-0366(20)30339-4.
[39] C.J. Jordan, Z.-X. Xi, Discovery and development of varenicline for smoking
cessation, Expert Opin. Drug Discov. 13 (7) (2018) 671683, https://doi.org/
10.1080/17460441.2018.1458090.
[40] Z. Justinova, G. Tanda, G.H. Redhi, S.R. Goldberg, Self-administration of delta9-
tetrahydrocannabinol (THC) by drug naive squirrel monkeys, Psychopharmacology
169 (2) (2003) 135140, https://doi.org/10.1007/s00213-003-1484-0.
[41] Z. Justinova, S.R. Goldberg, S.J. Heishman, G. Tanda, Self-administration of
cannabinoids by experimental animals and human marijuana smokers, Pharmacol.,
Biochem., Behav. 81 (2) (2005) 285299, https://doi.org/10.1016/j.
pbb.2005.01.026.
[42] Z. Justinova, P. Mascia, H.-Q. Wu, M.E. Secci, G.H. Redhi, L.V. Panlilio,
M. Scherma, C. Barnes, A. Parashos, T. Zara, W. Fratta, M. Solinas, M. Pistis,
J. Bergman, B.D. Kangas, S. Ferr´
e, G. Tanda, R. Schwarcz, S.R. Goldberg, Reducing
cannabinoid abuse and preventing relapse by enhancing endogenous brain levels of
kynurenic acid, Nat. Neurosci. 16 (11) (2013) 16521661, https://doi.org/
10.1038/nn.3540.
[43] I. Katona, E.A. Rancz, L. Acsady, C. Ledent, K. Mackie, N. Hajos, T.F. Freund,
Distribution of CB1 cannabinoid receptors in the amygdala and their role in the
control of GABAergic transmission, J. Neurosci.: Off. J. Soc. Neurosci. 21 (23)
(2001) 95069518, https://doi.org/10.1523/JNEUROSCI.21-23-09506.2001.
[44] B. Le Foll, B. Forget, H.-J. Aubin, S.R. Goldberg, Blocking cannabinoid CB1
receptors for the treatment of nicotine dependence: Insights from pre-clinical and
clinical studies, Addict. Biol. 13 (2) (2008) 239252, https://doi.org/10.1111/
j.1369-1600.2008.00113.x.
[45] C. L´
ena, A. de Kerchove dExaerde, M. Cordero-Erausquin, N. Le Nov`
ere, M. del
Mar Arroyo-Jimenez, J.-P. Changeux, Diversity and distribution of nicotinic
acetylcholine receptors in the locus ceruleus neurons, Proc. Natl. Acad. Sci. USA 96
(21) (1999) 1212612131.
[46] M. Liu, Y. Jiang, R. Wedow, Y. Li, D.M. Brazel, F. Chen, G. Datta, J. Davila-
Velderrain, D. McGuire, C. Tian, X. Zhan, 23andMe Research Team, HUNT All-In
Psychiatry, H. Choquet, A.R. Docherty, J.D. Faul, J.R. Foerster, L.G. Fritsche, M.
E. Gabrielsen, S. Vrieze, Association studies of up to 1.2 million individuals yield
new insights into the genetic etiology of tobacco and alcohol use, Nat. Genet. 51
(2) (2019) 237244, https://doi.org/10.1038/s41588-018-0307-5.
[47] C. Lopez-Quintero, J. P´
erez de los Cobos, D.S. Hasin, M. Okuda, S. Wang, B.
F. Grant, C. Blanco, Probability and predictors of transition from rst use to
dependence on nicotine, alcohol, cannabis, and cocaine: results of the National
Epidemiologic Survey on Alcohol and Related Conditions (NESARC), Drug Alcohol
Depend. 115 (12) (2011) 120130, https://doi.org/10.1016/j.
drugalcdep.2010.11.004.
[48] D.I. Lubman, A. Cheetham, M. Yücel, Cannabis and adolescent brain development,
Pharmacol. Ther. 148 (2015) 116, https://doi.org/10.1016/j.
pharmthera.2014.11.009.
[49] M.T. Lynskey, A. Agrawal, A. Henders, E.C. Nelson, P.A.F. Madden, N.G. Martin,
An australian twin study of cannabis and other illicit drug use and misuse, and
other psychopathology, Twin Res. Hum. Genet.: Off. J. Int. Soc. Twin Stud. 15 (5)
(2012) 631641, https://doi.org/10.1017/thg.2012.41.
[50] L.A. Manwell, T. Miladinovic, E. Raaphorst, S. Rana, S. Malecki, P.E. Mallet,
Chronic nicotine exposure attenuates the effects of Δ9-tetrahydrocannabinol on
anxiety-related behavior and social interaction in adult male and female rats, Brain
Behav. 9 (11) (2019), e01375, https://doi.org/10.1002/brb3.1375.
[51] G. Marsicano, B. Lutz, Expression of the cannabinoid receptor CB1 in distinct
neuronal subpopulations in the adult mouse forebrain, Eur. J. Neurosci. 11 (12)
(1999) 42134225, https://doi.org/10.1046/j.1460-9568.1999.00847.x.
[52] G. McKendrick, N.M. Graziane, Drug-induced conditioned place preference and its
practical use in substance use disorder research, Front. Behav. Neurosci. 14 (2020),
582147, https://doi.org/10.3389/fnbeh.2020.582147.
[53] A.L. McRae-Clark, K.M. Gray, N.L. Baker, B.J. Sherman, L. Squeglia, G.L. Sahlem,
A. Wagner, R. Tomko, Varenicline as a treatment for cannabis use disorder: a
placebo-controlled pilot trial, Drug Alcohol Depend. 229 (2021), 109111, https://
doi.org/10.1016/j.drugalcdep.2021.109111.
[54] G. Mereu, K.-W.P. Yoon, V. Boi, G.L. Gessa, L. Naes, T.C. Westfall, Preferential
stimulation of ventral tegmental area dopaminergic neurons by nicotine, Eur. J.
Pharmacol. 141 (3) (1987) 395399, https://doi.org/10.1016/0014-2999(87)
90556-5.
[55] M. Metna-Laurent, M. Mond´
esir, A. Grel, M. Vall´
ee, P.-V. Piazza, Cannabinoid-
induced tetrad in mice, Curr. Protoc. Neurosci. 80 (1) (2017) 9.59.19.59.10,
https://doi.org/10.1002/cpns.31.
[56] K.B. Mihalak, F.I. Carroll, C.W. Luetje, Varenicline is a partial agonist at
alpha4beta2 and a full agonist at alpha7 neuronal nicotinic receptors, Mol.
Pharmacol. 70 (3) (2006) 801805, https://doi.org/10.1124/mol.106.025130.
[57] T. Miladinovic, L.A. Manwell, E. Raaphorst, S.L. Malecki, S.A. Rana, P.E. Mallet,
Effects of chronic nicotine exposure on Δ9-tetrahydrocannabinol-induced
locomotor activity and neural activation in male and female adolescent and adult
rats, Pharmacol., Biochem., Behav. 194 (2020), 172931, https://doi.org/10.1016/
j.pbb.2020.172931.
[58] K. Monory, F. Massa, M. Egertov´
a, M. Eder, H. Blaudzun, R. Westenbroek,
W. Kelsch, W. Jacob, R. Marsch, M. Ekker, J. Long, J.L. Rubenstein, S. Goebbels, K.-
A. Nave, M. During, M. Klugmann, B. W¨
olfel, H.-U. Dodt, W. Zieglg¨
ansberger,
B. Lutz, The endocannabinoid system controls key epileptogenic circuits in the
hippocampus, Neuron 51 (4) (2006) 455466, https://doi.org/10.1016/j.
neuron.2006.07.006.
[59] P.P. Muldoon, A.H. Lichtman, L.H. Parsons, M.I. Damaj, The role of fatty acid
amide hydrolase inhibition in nicotine reward and dependence, Life Sci. 92 (89)
(2013) 458462, https://doi.org/10.1016/j.lfs.2012.05.015.
[60] D.A.L. Newcombe, The effect of varenicline administration on cannabis and
tobacco use in cannabis and nicotine dependent individuals ? A case-series,
J. Addict. Res. Ther. 06 (02) (2015), https://doi.org/10.4172/2155-
6105.1000222.
[61] L.R. Pacek, J. Copeland, L. Dierker, C.O. Cunningham, S.S. Martins, R.D. Goodwin,
Among whom is cigarette smoking declining in the United States? The impact of
cannabis use status, 20022015, Drug Alcohol Depend. 191 (2018) 355360,
https://doi.org/10.1016/j.drugalcdep.2018.01.040.
[62] L.V. Panlilio, S.R. Goldberg, Self-administration of drugs in animals and humans as
a model and an investigative tool, Addiction 102 (12) (2007) 18631870, https://
doi.org/10.1111/j.1360-0443.2007.02011.x.
[63] J. Patel, R. Marwaha, Cannabis Use Disorder. In StatPearls ([Internet]), StatPearls
Publishing, 2022 ([Internet]), https://www.ncbi.nlm.nih.gov/books/NBK
538131/.
[64] D.M. Penetar, A.R. Looby, E.T. Ryan, M.A. Maywalt, S.E. Lukas, Bupropion reduces
some of the symptoms of marihuana withdrawal in chronic marihuana users: a
pilot study, Subst. Abus.: Res. Treat. 6 (2012) 6371, https://doi.org/10.4137/
SART.S9706.
[65] F. Pistillo, F. Clementi, M. Zoli, C. Gotti, Nicotinic, glutamatergic and
dopaminergic synaptic transmission and plasticity in the mesocorticolimbic
system: focus on nicotine effects, Prog. Neurobiol. 124 (2015) 127, https://doi.
org/10.1016/j.pneurobio.2014.10.002.
[66] L. Ponzoni, M. Moretti, D. Braida, M. Zoli, F. Clementi, P. Viani, M. Sala, C. Gotti,
Increased sensitivity to Δ9-THC-induced rewarding effects after seven-week
exposure to electronic and tobacco cigarettes in mice, Eur.
Neuropsychopharmacol.: J. Eur. Coll. Neuropsychopharmacol. 29 (4) (2019)
566576, https://doi.org/10.1016/j.euroneuro.2019.02.001.
[67] R.A. Rabin, T.P. George, A review of co-morbid tobacco and cannabis use
disorders: possible mechanisms to explain high rates of co-use, Am. J. Addict. 24
(2) (2015) 105116, https://doi.org/10.1111/ajad.12186.
[68] D.E. Ramo, H. Liu, J.J. Prochaska, Tobacco and marijuana use among adolescents
and young adults: a systematic review of their co-use, Clin. Psychol. Rev. 32 (2)
(2012) 105121, https://doi.org/10.1016/j.cpr.2011.12.002.
[69] G.L. Ream, E. Benoit, B.D. Johnson, E. Dunlap, Smoking tobacco along with
marijuana increases symptoms of cannabis dependence, Drug Alcohol Depend. 95
(3) (2008) 199208, https://doi.org/10.1016/j.drugalcdep.2008.01.011.
[70] F. Rodríguez de Fonseca, I. Del Arco, F.J. Bermudez-Silva, A. Bilbao, A. Cippitelli,
M. Navarro, The endocannabinoid system: Physiology and pharmacology, Alcohol.
Alcohol. 40 (1) (2005) 214, https://doi.org/10.1093/alcalc/agh110.
[71] F. Rodrı guez de Fonseca, I. Del Arco, J.L. Martın-Calder´
on, M.A. Gorriti,
M. Navarro, Role of the endogenous cannabinoid system in the regulation of motor
activity, Neurobiol. Dis. 5 (6) (1998) 483501, https://doi.org/10.1006/
nbdi.1998.0217. Role of the Endogenous Cannabinoid System in the Regulation of
Motor Activity | Elsevier Enhanced Reader. (n.d.). https://doi.org/10.1006/
nbdi.1998.0217.
[72] R. Salas, R. Sturm, J. Boulter, M. De Biasi, Nicotinic receptors in the habenulo-
interpeduncular system are necessary for nicotine withdrawal in mice, J. Neurosci.:
Off. J. Soc. Neurosci. 29 (10) (2009) 30143018, https://doi.org/10.1523/
JNEUROSCI.4934-08.2009.
[73] G.L. Schauer, C.J. Berg, M.C. Kegler, D.M. Donovan, M. Windle, Differences in
tobacco product use among past month adult marijuana users and nonusers:
ndings from the 20032012 national survey on drug use and health, Nicotine Tob.
Res. 18 (3) (2016) 281288, https://doi.org/10.1093/ntr/ntv093.
[74] M.E. Secci, A. Auber, L.V. Panlilio, G.H. Redhi, E.B. Thorndike, C.W. Schindler,
R. Schwarcz, S.R. Goldberg, Z. Justinova, Attenuating nicotine reinforcement and
relapse by enhancing endogenous brain levels of kynurenic acid in rats and squirrel
monkeys, Neuropsychopharmacol.: Off. Publ. Am. Coll. Neuropsychopharmacol.
42 (8) (2017) 16191629, https://doi.org/10.1038/npp.2017.21.
[75] P. S´
egu´
ela, J. Wadiche, K. Dineley-Miller, J.A. Dani, J.W. Patrick, Molecular
cloning, functional properties, and distribution of rat brain alpha 7: a nicotinic
cation channel highly permeable to calcium, J. Neurosci.: Off. J. Soc. Neurosci. 13
(2) (1993) 596604, https://doi.org/10.1523/JNEUROSCI.13-02-00596.1993.
[76] I. Shim, H.-T. Kim, Y.-H. Kim, B.-G. Chun, D.-H. Hahm, E.H. Lee, S.E. Kim, H.-
J. Lee, Role of nitric oxide synthase inhibitors and NMDA receptor antagonist in
nicotine-induced behavioral sensitization in the rat, Eur. J. Pharmacol. 443 (13)
(2002) 119124, https://doi.org/10.1016/s0014-2999(02)01582-0.
[77] A.D. Smith, M.S. Dar, Mouse cerebellar nicotiniccholinergic receptor modulation
of Δ9-THC ataxia: Role of the
α
4β2 subtype, Brain Res. 1115 (1) (2006) 1625,
https://doi.org/10.1016/j.brainres.2006.07.075.
[78] A.D. Smith, M.S. Dar, Involvement of the
α
4β2 nicotinic receptor subtype in
nicotine-induced attenuation of Δ9-THC cerebellar ataxia: role of cerebellar nitric
oxide, Pharmacol. Biochem. Behav. 86 (1) (2007) 103112, https://doi.org/
10.1016/j.pbb.2006.12.013.
[79] M. Solinas, M. Scherma, L. Fattore, J. Stroik, C. Wertheim, G. Tanda, W. Fratta, S.
R. Goldberg, Nicotinic
α
7 receptors as a new target for treatment of cannabis abuse,
J. Neurosci. 27 (21) (2007) 56155620, https://doi.org/10.1523/
JNEUROSCI.0027-07.2007.
[80] M.S. Spano, P. Fadda, R. Frau, L. Fattore, W. Fratta, Cannabinoid self-
administration attenuates PCP-induced schizophrenia-like symptoms in adult rats,
B. Buzzi et al.
Pharmacological Research 191 (2023) 106746
8
Eur. Neuropsychopharmacol. 20 (1) (2010) 2536, https://doi.org/10.1016/j.
euroneuro.2009.09.004.
[81] S. Spencer, D. Neuhofer, V. Chioma, C. Garcia-Keller, D. Schwartz, N. Allen,
M. Scoeld, T. Ortiz-Ithier, P.W. Kalivas, A model of Δ9-tetrahydrocannabinol
(THC) self-administration and reinstatement that alters synaptic plasticity in
nucleus accumbens, Biol. Psychiatry 84 (8) (2018) 601610, https://doi.org/
10.1016/j.biopsych.2018.04.016.
[82] S.J. Stringeld, B.E. Sanders, J.A. Suppo, A.F. Sved, M.M. Torregrossa, Nicotine
enhances intravenous self-administration of cannabinoids in adult rats, Nicotine
Tob. Res.: Off. J. Soc. Res. Nicotine Tob. (2022) ntac267, https://doi.org/10.1093/
ntr/ntac267.
[83] G. Tanda, P. Munzar, S.R. Goldberg, Self-administration behavior is maintained by
the psychoactive ingredient of marijuana in squirrel monkeys, Nat. Neurosci. 3
(11) (2000) 10731074, https://doi.org/10.1038/80577.
[84] J.R. Tregellas, K.P. Wylie, Alpha7 nicotinic receptors as therapeutic targets in
schizophrenia, Nicotine Tob. Res.: Off. J. Soc. Res. Nicotine Tob. 21 (3) (2019)
349356, https://doi.org/10.1093/ntr/nty034.
[85] E. Valjent, J.M. Mitchell, M.-J. Besson, J. Caboche, R. Maldonado, Behavioural and
biochemical evidence for interactions between Δ9-tetrahydrocannabinol and
nicotine, Br. J. Pharmacol. 135 (2) (2002) 564578, https://doi.org/10.1038/sj.
bjp.0704479.
[86] V. Vengeliene, N. Cannella, T. Takahashi, R. Spanagel, Metabolic shift of the
kynurenine pathway impairs alcohol and cocaine seeking and relapse,
Psychopharmacology 233 (18) (2016) 34493459, https://doi.org/10.1007/
s00213-016-4384-9.
[87] K.J.H. Verweij, J.M. Vink, A. Abdellaoui, N.A. Gillespie, E.M. Derks, J.L. Treur, The
genetic aetiology of cannabis use: from twin models to genome-wide association
studies and beyond (Article), Transl. Psychiatry 12 (1) (2022) 1, https://doi.org/
10.1038/s41398-022-02215-2.
[88] K.J.H. Verweij, B.P. Zietsch, M.T. Lynskey, S.E. Medland, M.C. Neale, N.G. Martin,
D.I. Boomsma, J.M. Vink, Genetic and environmental inuences on cannabis use
initiation and problematic use: a meta-analysis of twin studies, Addiction 105 (3)
(2010) 417430, https://doi.org/10.1111/j.1360-0443.2009.02831.x.
[89] A.G.P. Wakeford, B.B. Wetzell, R.L. Pomfrey, M.M. Clasen, W.W. Taylor, B.
J. Hempel, A.L. Riley, The effects of cannabidiol (CBD) on Δ
9
-
tetrahydrocannabinol (THC) self-administration in male and female Long-Evans
rats, Exp. Clin. Psychopharmacol. 25 (2017) 242248, https://doi.org/10.1037/
pha0000135.
[90] C.T. Whitlow, C.S. Freedland, L.J. Porrino, Functional consequences of the
repeated administration of Delta9-tetrahydrocannabinol in the rat, Drug Alcohol
Depend. 71 (2) (2003) 169177, https://doi.org/10.1016/s0376-8716(03)00135-
2.
[91] S. Wilkes, The use of bupropion SR in cigarette smoking cessation, Int. J. Chronic
Obstr. Pulm. Dis. 3 (1) (2008) 4553, https://doi.org/10.2147/copd.s1121.
[92] R.E. Wittenberg, S.L. Wolfman, M. De Biasi, J.A. Dani, Nicotinic acetylcholine
receptors and nicotine addiction: a brief introduction, Neuropharmacology 177
(2020), 108256, https://doi.org/10.1016/j.neuropharm.2020.108256.
[93] World Drug Report 2021. (n.d.). United Nations: Ofce on Drugs and Crime.
Retrieved December 13, 2022, from //www.unodc.org/unodc/en/data-and-
analysis/wdr2021.html.
[94] R. Young, Drug discrimination, in: J.J. Buccafusco (Ed.), Methods of Behavior
Analysis in Neuroscience, second ed.., CRC Press/Taylor & Francis, 2009. http:
//www.ncbi.nlm.nih.gov/books/NBK5225/.
B. Buzzi et al.
... Cannabis and nAChR receptors codistribute in the same brain areas, suggesting that the two systems can engage in crosstalk [86]. Tobacco and cannabis are the most common drugs of abuse consumed by adolescents and young adults [87,88]. The co-use of these drugs has been suggested to produce mutually reinforcing effects and a decrease in adverse effects [89]. ...
... A recent study found that individuals with cannabinoid use disorder present reduced expression of the CHRNA2 gene in the cerebellum, suggesting that the gene that encodes for the α2 nAChR subunit may be involved in the susceptibility to developing this disorder. Furthermore, a negative correlation between the gene expressions of CHRNA2 and CNR1 (cannabinoid receptor 1) in the cerebellar cortex and cerebellar nuclei has been reported [87]. Participation of the homomeric ɑ7 nAChR has been linked to the rewarding effects of cannabinoid use, while the ɑ4β2 nAChR subtype has been associated with a reduction in cannabinoid-induced ataxia, and as such with a reduction in cannabinoid-induced motor impairment. ...
... Participation of the homomeric ɑ7 nAChR has been linked to the rewarding effects of cannabinoid use, while the ɑ4β2 nAChR subtype has been associated with a reduction in cannabinoid-induced ataxia, and as such with a reduction in cannabinoid-induced motor impairment. In addition, the potential roles of the ɑ5, ɑ3, and β4 nAChR subunits in cannabinoid use disorder, particularly in tolerance-and withdrawal-associated symptoms, have been addressed [87]. ...
Article
Full-text available
The cholinergic system plays an essential role in brain development, physiology, and pathophysiology. Herein, we review how specific alterations in this system, through genetic mutations or abnormal receptor function, can lead to aberrant neural circuitry that triggers disease. The review focuses on the nicotinic acetylcholine receptor (nAChR) and its role in addiction and in neurodegenerative and neuropsychiatric diseases and epilepsy. Cholinergic dysfunction is associated with inflammatory processes mainly through the involvement of α7 nAChRs expressed in brain and in peripheral immune cells. Evidence suggests that these neuroinflammatory processes trigger and aggravate pathological states. We discuss the preclinical evidence demonstrating the therapeutic potential of nAChR ligands in Alzheimer disease, Parkinson disease, schizophrenia spectrum disorders, and in autosomal dominant sleep-related hypermotor epilepsy. PubMed and Google Scholar bibliographic databases were searched with the keywords indicated below.
... In addition to their well-established role in tobacco smoking and other forms of nicotine dependence, nAChRs have also been implicated in the pathophysiology of other types of addiction and investigated as possible treatment targets. Buzzi et al. (2023) [23] review studies showing the important role played by various nAChR subtypes in cannabinoid-dependent reward, dependence and withdrawal, and the promising results of trials of nicotinic drugs for the treatment of cannabinoid use disorder. Kamens et al. (2023) [24] summarise the genetic and pharmacological data indicating the involvement of nAChRs in some of the behavioural effects of alcohol, including reward and sedation, and the efficacy of nicotinic drugs in decreasing alcohol consumption. ...
... In addition to their well-established role in tobacco smoking and other forms of nicotine dependence, nAChRs have also been implicated in the pathophysiology of other types of addiction and investigated as possible treatment targets. Buzzi et al. (2023) [23] review studies showing the important role played by various nAChR subtypes in cannabinoid-dependent reward, dependence and withdrawal, and the promising results of trials of nicotinic drugs for the treatment of cannabinoid use disorder. Kamens et al. (2023) [24] summarise the genetic and pharmacological data indicating the involvement of nAChRs in some of the behavioural effects of alcohol, including reward and sedation, and the efficacy of nicotinic drugs in decreasing alcohol consumption. ...
... CHRNA2 encodes for the α2 subunit of neuronal acetylcholine receptors, which are important for the binding of neurotransmitters. Scientists found that neuronal acetylcholine receptors modulate the withdrawal of THC, which is the main active ingredient in cannabis (Buzzi et al., 2023), thus playing a major role in cannabis use disorder susceptibility. Specifically, low levels of this α2 subunit, CHRNA2, in the cerebellum is associated with increased risk for cannabis use disorder. ...
Article
Abstracts Introduction The present study aimed to assess the efficacy of folic acid (FA) on withdrawal following nicotine (Nic) administration in adolescent male rats. Methods Adolescent male rats were divided into two groups: 1) vehicle and 2)Nic (Nic-2mg/kg), and were under the treatment from 21 to 42 days of age. After that, they continued the experiment without treatment and returned to a regular diet, except for one of those who received Nic. The rats were divided into four groups where they were treated with different doses of FA (5, 10, and 15 mg/kg) and bupropion (Bup) by oral gavage, and the final group included normal rats that received only FA (15mg/kg) from 42 days of age for three weeks during which withdrawal occurred. Results Results showed that adolescent Nic exposure exacerbated the behavioral indices of anxiety- and depression-like behaviors, while FA attenuated the effects of Nic withdrawal on anxiety and depression as well as Bup. In support, the biochemical results demonstrated a balance between oxidant and antioxidant mediators in addition to increase and decrease of serotonin and monoamine oxidase (MAO) activity in cortical tissue. TNF-α as an inflammatory agent was decreased, whereas IL-10 as an anti-inflammatory parameter was increased. Conclusion The present findings suggest anxiety and depression caused by Nic withdrawal were attenuated by FA more likely through reduction activity of MAO, the important enzyme responsible for serotonin metabolism along with balance between oxidant/anti-oxidant and pro-inflammatory/anti-inflammatory mediators. However, various mechanisms might be involved, which requires further investigation. Implications Nic withdrawal induced depression and anxiety like behavior in rats followed by neuro-oxidative damage and neuro-inflammation. Folic acid supplementation as well as bupropion improved cognitive disorders induced by Nic withdrawal by increasing neuro-inflammation, neuro-oxidative damage.
Article
Full-text available
The detrimental physical, mental, and socioeconomic effects of substance use disorders (SUDs) have been apparent to the medical community for decades. However, it has become increasingly urgent in recent years to develop novel pharmacotherapies to treat SUDs. Currently, practitioners typically rely on monotherapy. Monotherapy has been shown to be superior to no treatment at all for most substance classes. However, many randomized controlled trials (RCTs) have revealed that monotherapy leads to poorer outcomes when compared with combination treatment in all specialties of medicine. The results of RCTs suggest that monotherapy frequently fails since multiple dysregulated pathways, enzymes, neurotransmitters, and receptors are involved in the pathophysiology of SUDs. As such, research is urgently needed to determine how various neurobiological mechanisms can be targeted by novel combination treatments to create increasingly specific yet exceedingly comprehensive approaches to SUD treatment. This article aims to review the neurobiology that integrates many pathophysiologic mechanisms and discuss integrative pharmacology developments that may ultimately improve clinical outcomes for patients with SUDs. Many neurobiological mechanisms are known to be involved in SUDs. These include dopaminergic, nicotinic, N-methyl-D-aspartate (NMDA), and kynurenic acid (KYNA) mechanisms, for example. Emerging evidence indicates that KYNA, a tryptophan metabolite, modulates all these major pathophysiologic mechanisms. Therefore, achieving KYNA homeostasis by harmonizing integrative pathophysiology and pharmacology could prove to be a better therapeutic approach for SUDs. We propose KYNA-NMDA-α7nAChRcentric pathophysiology, the “conductor of the orchestra,” as a novel approach to treat many SUDs concurrently. KYNA-NMDA-7nAChR pathophysiology may be the “command center” of neuropsychiatry. To date, extant RCTs have shown equivocal findings across comparison conditions, possibly because investigators targeted single pathophysiologic mechanisms, hit wrong targets in underlying pathophysiologic mechanisms, and tested inadequate monotherapy treatment. We provide examples of potential combination treatments that simultaneously target multiple pathophysiologic mechanisms in addition to KYNA. Kynurenine pathway metabolism demonstrates the greatest potential as a target for neuropsychiatric diseases. The investigational medications with the most evidence include memantine, galantamine, and N-acetylcysteine. Future RCTs are warranted with novel combination treatments for SUDs. Multicenter RCTs with integrative pharmacology offer a promising, potentially fruitful avenue to develop novel therapeutics for the treatment of SUDs.
Article
Full-text available
Cannabis is among the most widely consumed psychoactive substances worldwide. Individual differences in cannabis use phenotypes can partly be explained by genetic differences. Technical and methodological advances have increased our understanding of the genetic aetiology of cannabis use. This narrative review discusses the genetic literature on cannabis use, covering twin, linkage, and candidate-gene studies, and the more recent genome-wide association studies (GWASs), as well as the interplay between genetic and environmental factors. Not only do we focus on the insights that these methods have provided on the genetic aetiology of cannabis use, but also on how they have helped to clarify the relationship between cannabis use and co-occurring traits, such as the use of other substances and mental health disorders. Twin studies have shown that cannabis use is moderately heritable, with higher heritability estimates for more severe phases of use. Linkage and candidate-gene studies have been largely unsuccessful, while GWASs so far only explain a small portion of the heritability. Dozens of genetic variants predictive of cannabis use have been identified, located in genes such as CADM2, FOXP2, and CHRNA2. Studies that applied multivariate methods (twin models, genetic correlation analysis, polygenic score analysis, genomic structural equation modelling, Mendelian randomisation) indicate that there is considerable genetic overlap between cannabis use and other traits (especially other substances and externalising disorders) and some evidence for causal relationships (most convincingly for schizophrenia). We end our review by discussing implications of these findings and suggestions for future work.
Article
Full-text available
The conditioned place preference (CPP) paradigm is a well-established model utilized to study the role of context associations in reward-related behaviors, including both natural rewards and drugs of abuse. In this review article, we discuss the basic history, various uses, and considerations that are tied to this technique. There are many potential takeaway implications of this model, including negative affective states, conditioned drug effects, memory, and motivation, which are all considered here. We also discuss the neurobiology of CPP including relevant brain regions, molecular signaling cascades, and neuromodulatory systems. We further examine some of our prior findings and how they integrate CPP with self-administration paradigms. Overall, by describing the fundamentals of CPP, findings from the past few decades, and implications of using CPP as a research paradigm, we have endeavored to support the case that the CPP method is specifically advantageous for studying the role of a form of Pavlovian learning that associates drug use with the surrounding environment.
Article
Full-text available
Background Variation in liability to cannabis use disorder has a strong genetic component (estimated twin and family heritability about 50–70%) and is associated with negative outcomes, including increased risk of psychopathology. The aim of the study was to conduct a large genome-wide association study (GWAS) to identify novel genetic variants associated with cannabis use disorder. Methods To conduct this GWAS meta-analysis of cannabis use disorder and identify associations with genetic loci, we used samples from the Psychiatric Genomics Consortium Substance Use Disorders working group, iPSYCH, and deCODE (20 916 case samples, 363 116 control samples in total), contrasting cannabis use disorder cases with controls. To examine the genetic overlap between cannabis use disorder and 22 traits of interest (chosen because of previously published phenotypic correlations [eg, psychiatric disorders] or hypothesised associations [eg, chronotype] with cannabis use disorder), we used linkage disequilibrium score regression to calculate genetic correlations. Findings We identified two genome-wide significant loci: a novel chromosome 7 locus (FOXP2, lead single-nucleotide polymorphism [SNP] rs7783012; odds ratio [OR] 1·11, 95% CI 1·07–1·15, p=1·84 × 10⁻⁹) and the previously identified chromosome 8 locus (near CHRNA2 and EPHX2, lead SNP rs4732724; OR 0·89, 95% CI 0·86–0·93, p=6·46 × 10⁻⁹). Cannabis use disorder and cannabis use were genetically correlated (rg 0·50, p=1·50 × 10⁻²¹), but they showed significantly different genetic correlations with 12 of the 22 traits we tested, suggesting at least partially different genetic underpinnings of cannabis use and cannabis use disorder. Cannabis use disorder was positively genetically correlated with other psychopathology, including ADHD, major depression, and schizophrenia. Interpretation These findings support the theory that cannabis use disorder has shared genetic liability with other psychopathology, and there is a distinction between genetic liability to cannabis use and cannabis use disorder. Funding National Institute of Mental Health; National Institute on Alcohol Abuse and Alcoholism; National Institute on Drug Abuse; Center for Genomics and Personalized Medicine and the Centre for Integrative Sequencing; The European Commission, Horizon 2020; National Institute of Child Health and Human Development; Health Research Council of New Zealand; National Institute on Aging; Wellcome Trust Case Control Consortium; UK Research and Innovation Medical Research Council (UKRI MRC); The Brain & Behavior Research Foundation; National Institute on Deafness and Other Communication Disorders; Substance Abuse and Mental Health Services Administration (SAMHSA); National Institute of Biomedical Imaging and Bioengineering; National Health and Medical Research Council (NHMRC) Australia; Tobacco-Related Disease Research Program of the University of California; Families for Borderline Personality Disorder Research (Beth and Rob Elliott) 2018 NARSAD Young Investigator Grant; The National Child Health Research Foundation (Cure Kids); The Canterbury Medical Research Foundation; The New Zealand Lottery Grants Board; The University of Otago; The Carney Centre for Pharmacogenomics; The James Hume Bequest Fund; National Institutes of Health: Genes, Environment and Health Initiative; National Institutes of Health; National Cancer Institute; The William T Grant Foundation; Australian Research Council; The Virginia Tobacco Settlement Foundation; The VISN 1 and VISN 4 Mental Illness Research, Education, and Clinical Centers of the US Department of Veterans Affairs; The 5th Framework Programme (FP-5) GenomEUtwin Project; The Lundbeck Foundation; NIH-funded Shared Instrumentation Grant S10RR025141; Clinical Translational Sciences Award grants; National Institute of Neurological Disorders and Stroke; National Heart, Lung, and Blood Institute; National Institute of General Medical Sciences.
Article
Introduction Nicotine and cannabis are commonly used together, yet few studies have investigated the effects of concurrent administration. Nicotine exhibits reinforcement enhancing effects by promoting the reinforcing properties of stimuli including other drugs. As many studies of this effect used non-contingent nicotine, we implemented a dual-self-administration model where rats have simultaneous access to two drugs and choose which to self-administer throughout a session. Here, we investigated the effect of self-administered or non-contingently delivered nicotine on cannabinoid self-administration. Methods Adult male rats were allowed to self-administer the synthetic cannabinoid WIN 55,212-2 (WIN) intravenously, with or without subcutaneous nicotine injections before each session. A separate group of animals were allowed to self-administer WIN, nicotine, or saline using a dual-catheter procedure, where each solution was infused independently and associated with a separate operant response. A third group of male and female rats were allowed to self-administer delta-9-tetrahydrocannabinol (THC) with or without pre-session injections of nicotine. Results Nicotine injections increased self-administration of WIN and THC. During dual self-administration, nicotine availability increased saline and WIN infusions but nicotine intake was not changed by WIN or saline availability. Rats preferred nicotine over saline, but preferred nicotine and WIN equally when both were available. The effect of nicotine on cannabinoid self-administration was acute and reversible when nicotine was no longer present. Conclusions These results expand our understanding of the ability of nicotine to enhance reinforcement of other drugs and suggest that co-use of nicotine and cannabinoids promotes cannabinoid use beyond what would be taken alone. Implications This study utilizes a dual intravenous self-administration model to investigate the ability of nicotine to enhance cannabinoid intake. Our results demonstrate that the reinforcement enhancing properties of nicotine on drug use extend to include cannabinoids, but that this effect occurs specifically when nicotine is administered alongside the cannabinoid. Interestingly, cannabinoid use did not promote nicotine intake, suggesting this mechanism of reinforcement is specific to nicotine.
Article
Background: Cannabis is among the most consumed psychoactive substances world-wide. Considering changing policy trends regarding the substance, it is crucial to understand more clearly its potential acute and residual adverse effects from a public health viewpoint. Cognitive function is one of the targeted areas with conflicting findings. This meta-review measured the magnitude of acute and residual effects of cannabis on cognition in adolescents and adults provided by meta-analyses and evaluated quality of evidence. Methods: A systematic search was performed in PubMed, PsycINFO, Web of Science and Google Scholar. Meta-analyses were included if they quantitatively examined the performances of users from the general population on cognitive tasks. Results: The search retrieved 10 eligible meta-analyses (71 effects sizes, n = 43 761) with evidence ranging from low to moderate quality, which were categorized into domains of cognitive functions: executive functions (k = 7), learning and memory (k = 5), attention (k = 4), processing speed (k = 5), perceptual motor function (k = 2) and language (k = 2). Verbal learning and memory displayed the most robust evidence and were most impaired by acute cannabis intoxication that persisted after intoxication passed. Small-to-moderate acute and residual adverse effects were reported for executive functioning. Cannabis use led to small deficits in inhibitory processes and flexibility, whereas small-to-moderate deficits were reported for working memory and decision-making. Evidence regarding processing speed and attention has shown that cannabis administration induced small-to-moderate adverse effects and residual neurocognitive deficits were observed in heavy cannabis-using youths. Results showed no significant difference between cannabis users and non-users on language, and small-to-moderate effects for simple motor skills. Conclusion: Meta-analytical data on the acute effects of cannabis use on neurocognitive function have shown that cannabis intoxication leads to small to moderate deficits in several cognitive domains. These acute impairments accord with documented residual effects, suggesting that the detrimental effects of cannabis persist beyond acute intake.
Chapter
The prevalence of cannabis abuse and dependence has been increasing among both adolescents and adults in the United States. This chapter reviews the reviews psychosocial consequences associated with cannabis use, assessing for cannabis use disorder, and treatment options.
Article
Background An efficacious pharmacotherapy for cannabis use disorder (CUD) has yet to be established. This study preliminarily evaluated the safety and efficacy of varenicline for CUD in a proof-of-concept clinical trial. Methods Participants in this 6-week randomized, placebo-controlled pilot trial received either varenicline (n=35) or placebo (n=37), added to a brief motivational enhancement therapy intervention. Outcomes included cannabis withdrawal, cannabis abstinence, urine cannabinoid levels, percent cannabis use days, and cannabis sessions per day. Results Both treatment groups noted significant decreases in self-reported cannabis withdrawal, percentage of days used, and use sessions per day during treatment compared to baseline. While this pilot trial was not powered to detect statistically significant between-group differences, participants randomized to varenicline evidenced numerically greater rates of self-reported abstinence at the final study visit [Week 6 intent-to-treat (ITT): Varenicline: 17.1% vs. Placebo: 5.4%; RR=3.2 (95% CI: 0.7,14.7)]. End-of-treatment urine creatinine corrected cannabinoid levels were numerically lower in the varenicline group and higher in the placebo group compared to baseline [Change from baseline: Varenicline -1.7 ng/mg (95% CI: -4.1,0.8) vs. Placebo: 1.9 ng/mg (95% CI: -0.4,4.3); Δ=3.5 (95% CI: 0.1,6.9)]. Adverse events related to study treatment did not reveal new safety signals. Conclusions Findings support the feasibility of conducting clinical trials of varenicline as a candidate pharmacotherapy for CUD, and indicate that a full-scale efficacy trial, powered based on effect sizes and variability yielded in this study, is warranted.
Article
Nicotine is a highly addictive drug found in tobacco that drives its continued use despite the harmful consequences. The initiation of nicotine abuse involves the mesolimbic dopamine system, which contributes to the rewarding sensory stimuli and associative learning processes in the beginning stages of addiction. Nicotine binds to neuronal nicotinic acetylcholine receptors (nAChRs), which come in a diverse collection of subtypes. The nAChRs that contain the α4 and β2 subunits, often in combination with the α6 subunit, are particularly important for nicotine's ability to increase midbrain dopamine neuron firing rates and phasic burst firing. Chronic nicotine exposure results in numerous neuroadaptations, including the upregulation of particular nAChR subtypes associated with long-term desensitization of the receptors. When nicotine is no longer present, for example during attempts to quit smoking, a withdrawal syndrome develops. The expression of physical withdrawal symptoms depends mainly on the α2, α3, α5, and β4 nicotinic subunits in the epithalamic habenular complex and its target regions. Thus, nicotine affects diverse neural systems and an array of nAChR subtypes to mediate the overall addiction process. This article is part of the special issue on ‘Contemporary Advances in Nicotine Neuropharmacology’.
Article
Rationale High rates of comorbid tobacco and cannabis use in adolescents and young adults may be related to functional interactions between the nicotinic cholinergic and cannabinoid systems in the brain during development. This study examined the effects of chronic exposure to nicotine (the psychoactive component in tobacco) on acute exposure to delta-9-tetrahydrocannabinol (THC) (the psychoactive component of cannabis). Methods Male and female adolescent and adult Sprague-Dawley rats (N = 112) were injected daily with nicotine (1 mg/kg, i.p.) or vehicle for 14 days, followed by a 14-day drug-free period. On test day, rats were injected with THC (5 mg/kg, i.p.) or vehicle, locomotor activity was recorded for 2 h, and brains harvested for c-Fos immunoreactivity (IR). Results Locomotor activity and c-Fos IR changes induced by THC challenge were altered by nicotine pre-exposure and modified by age and sex. THC-induced suppression of locomotor activity was attenuated by nicotine pre-exposure in adult but not adolescent males. THC-induced suppression of locomotor activity was potentiated by nicotine pre-exposure in female adolescents, with no effects of THC or nicotine observed in female adults. THC increased c-Fos IR in the caudate, nucleus accumbens, stria terminalis, septum, amygdala, hypothalamus, and thalamus. Nicotine pre-exposure potentiated this effect in all regions. Several brain regions showed age and sex differences in c-Fos IR such that expression was greater in adults than adolescents and in females than males. Conclusions Chronic nicotine pre-exposure produces lasting effects on cannabinoid-mediated signalling in the brain and on behaviour that are mediated by age and sex. Funding support NSERC.