Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Feb 19.
Published in final edited form as: Annu Rev Neurosci. 2016 Feb 24;39:1–17. doi: 10.1146/annurev-neuro-070815-014038

Beyond the CB1 Receptor: Is Cannabidiol the Answer for Disorders of Motivation?

Natalie E Zlebnik 1, Joseph F Cheer 1,2
PMCID: PMC5818147  NIHMSID: NIHMS942303  PMID: 27023732

Abstract

The Cannabis sativa plant has been used to treat various physiological and psychiatric conditions for millennia. Current research is focused on isolating potentially therapeutic chemical constituents from the plant for use in the treatment of many central nervous system disorders. Of particular interest is the primary nonpsychoactive constituent cannabidiol (CBD). Unlike Δ9-tetrahydrocannabinol (THC), CBD does not act through the cannabinoid type 1 (CB1) receptor but has many other receptor targets that may play a role in psychiatric disorders. Here we review preclinical and clinical data outlining the therapeutic efficacy of CBD for the treatment of motivational disorders such as drug addiction, anxiety, and depression. Across studies, findings suggest promising treatment effects and potentially overlapping mechanisms of action for CBD in these disorders and indicate the need for further systematic investigation of the viability of CBD as a psychiatric pharmacotherapy.

Keywords: cannabidiol, Δ9-tetrahydrocannabinol, THC, reward, addiction, anxiety, depression

INTRODUCTION

Since antiquity, various preparations of the plant Cannabis sativa have been used for medicinal and religious purposes. Widespread across ancient Asia, cannabis served as an antiemetic, anticonvulsant, antibiotic, anti-inflammatory, analgesic, anesthetic, antispasmodic, diuretic, digestive, appetite stimulant, antitussive, expectorant, and aphrodisiac (Zuardi 2006). With origins as far back as 4000 BC, Chinese pharmacopoeias documented the medicinal use of cannabis, outlining its effectiveness in the treatment of pain, constipation, menstrual cramps, and malaria (Li 1978). However, these sources noted that, when taken in excess, cannabis could yield “visions of devils” (Li 1978, p. 18). Its psychoactive properties were also described in ancient India, where they were associated with religious rituals, and the sacred text Atharva Veda included cannabis as one of the five sacred plants for its ability to convey “happiness,” “joy,” and “freedom” (Touw 1981, p. 25). Additionally, it is well documented that people used cannabis to alleviate symptoms of mood disorders such as anxiety, depression, mania, and hysteria (Mechoulam et al. 1970, Russo & Guy 2006).

Medical interest in the plant spread throughout Europe and North America in the nineteenth century (Grinspoon & Bakalar 1995), but despite the apparent therapeutic potential of cannabis for affective disorders and innumerous other ailments, its use in Western medicine decreased significantly in the early twentieth century (Zuardi 2006). The decline of medicinal cannabis was largely due to several factors. In particular, researchers may have had difficulty in achieving reliable therapeutic effects, as drug extracts were made from different strains of the plant and with varying methods of preparation (Fankhauser 2002). Because the active constituents of cannabis had not been isolated, other medications replaced cannabis for its recommended uses, and it was eventually removed from Western pharmacopoeias. Finally, concerns regarding the psychoactive effects of cannabis and its potential impairment of learning and memory prompted many legal restrictions, obstructing academic research on the plant (Zuardi 2006, Hill et al. 2012).

Systematic quantitative research on the active constituents of cannabis did not begin in earnest until modern separation techniques made it possible to distinguish among the closely related molecular compounds within the plant (Mechoulam & Parker 2013). Although scientists eventually isolated over 60 phytocannabinoids (pCBs), the two major active constituents identified were Δ9-tetrahydrocannabinol (THC) (Gaoni & Mechoulam 1964) and cannabidiol (CBD) (Mechoulam & Shvo 1963). THC is the prominent psychoactive pCB of the plant and mediates the rewarding properties of cannabis (Huestis et al. 2001); CBD, in contrast, does not have reinforcing effects and has low abuse potential (Katsidoni et al. 2013, Parker et al. 2004). Despite these differences in psychoactivity, THC and CBD are produced by codominant alleles of the same gene locus and segregate according to simple Mendelian inheritance, yielding three cannabis chemovariants in a 1:2:1 ratio of pure THC, mixed THC/CBD, and pure CBD (de Meijer et al. 2003). Currently, although the chemovariants containing predominantly THC are used primarily for recreational purposes, synthetic THC (e.g., dronabinol, nabilone) has been approved for the alleviation of nausea and vomiting as well as for appetite stimulation in cancer and HIV/AIDS patients (Hill et al. 2012). Perhaps of greater therapeutic interest are the chemovariants containing a 1:1 ratio of THC to CBD or those containing predominantly CBD. As the first licensed medicinal whole cannabis extract [in Canada, the United Kingdom (UK), Germany, Spain, Denmark, and New Zealand], Sativex® (GW Pharmaceuticals, UK) has indications for pain and spasticity associated with multiple sclerosis (Barnes 2006, Perras 2005) and an approximate 1:1 ratio of THC to CBD. Importantly, this ratio reduces the unwanted central actions of THC (Russo & Guy 2006) and minimizes abuse liability (Schoedel et al. 2011). This has renewed interest in investigation of synergistic or “entourage” effects of pCBs administered together (Ben-Shabat et al. 1998, Mechoulam & Ben-Shabat 1999) and highlighted the need for systematic examination of pCBs other than THC.

Although CBD is the second most common pCB, there has been a shortage of experimental inquiry into its mechanisms of action and therapeutic effects. When administered along with THC, CBD reduces subjective ratings of intoxication (Robson 2011, Schoedel et al. 2011), ameliorates cognitive and behavioral impairment (Wade et al. 2004), and reverses THC-induced anxiety (Zuardi et al. 1982). However, CBD might be worth investigating not just for its ability to antagonize the effects of THC but in its own right as a pharmacological agent. Early preclinical studies demonstrate promising treatment effects of CBD for seizure disorders (Devinsky et al. 2014), and along with the development of CBD-based pharmacotherapies (e.g., Epidiolex®, GW Pharmaceuticals), clinical trials are under way to evaluate the strength of its treatment effects for medically intractable pediatric epilepsy (Cilio et al. 2014, Devinsky et al. 2014). The potential for wide-ranging therapeutic action of CBD in central nervous system disorders extends from emerging evidence of its diversity of receptor targets, including those beyond the endocannabinoid (eCB) system. Here, we review the pharmacological actions of CBD that may make it suitable as a treatment for an array of motivational and affective disorders such as drug addiction, anxiety, and depression.

BRIEF OVERVIEW OF THE ENDOCANNABINOID SYSTEM

Following the isolation of THC from cannabis (Gaoni & Mechoulam 1964), researchers discovered that it binds to two different G protein–coupled receptors (GPCRs), the cannabinoid type 1 (CB1) (Devane et al. 1988) and CB2 (Munro et al. 1993) receptors. Whereas the CB2 receptor is distributed primarily in immune cells, the CB1 receptor is found throughout the CNS (Herkenham et al. 1990). It is the most abundant GPCR in the brain (Mechoulam & Parker 2013), and consistent with its role in reward and cognition, its highest densities are in the basal ganglia, substantia nigra, globus pallidus, hippocampus, and cerebellum (Herkenham et al. 1990). Sharing 48% sequence homology, both receptors couple to inhibitory Gi/Go proteins and have the ability to activate several signal transduction mechanisms to inhibit voltage-gated calcium channels and adenylyl cyclase as well as activate potassium channels and MAP kinase (Bidaut-Russell et al. 1990, Henry & Chavkin 1995, Hoffman & Lupica 2000, Twitchell et al. 1997). The endogenous ligands N-arachidonoylethanolamine (AEA) or anandamide (from the Sanskrit word ananda, meaning bliss) (Devane et al. 1992) and 2-arachidonoylglycerol (2-AG) (Mechoulam et al. 1995, Sugiura et al. 1995) were identified shortly thereafter. Unlike classical neurotransmitters, eCBs function as retrograde messengers in several brain regions, including the nucleus accumbens (NAc) and ventral tegmental area (VTA) (Melis et al. 2004, Robbe et al. 2002, Wilson & Nicoll 2001); they are not stored in vesicles but rather are synthesized on demand within the postsynaptic cell in response to large increases in intracellular calcium levels during periods of sustained neural activation (Freund et al. 2003). Once synthesized, they cross the plasma membrane and activate CB1 receptors on presynaptic terminals, thereby inhibiting neurotransmitter release (Wilson & Nicoll 2001). Thus, the retrograde action of eCBs permits postsynaptic neurons to adjust their synaptic input as a function of their relative level of activation and may play an important role in regulating synaptic plasticity (Alger 2009).

PHYTOCANNABINOIDS AND DISORDERS OF MOTIVATION: MECHANISMS OF ACTION

Δ9-Tetrahydrocannabinol

Self-administration of THC ( Justinova et al. 2008) and the positive subjective effects of smoked marijuana (Huestis et al. 2001) are blocked by administration of the CB1 receptor inverse agonist rimonabant, suggesting that the characteristic reinforcing or pleasurable effects of cannabis are mediated by the CB1 receptor. THC is a partial agonist of the CB1 receptor (Pertwee 2008), and similar to most other drugs of abuse, it modulates the mesolimbic dopamine system. This neural pathway is composed of dopamine cells originating in the VTA of the midbrain that project rostrally to the NAc in the ventral forebrain and to the prefrontal cortex (Wise & Bozarth 1985). The rewarding properties of all known drugs of abuse are thought to arise from dopamine release in the NAc (Di Chiara & Imperato 1988, Roberts et al. 1977, Volkow & Morales 2015, Wise & Bozarth 1985). Importantly, administration of THC or cannabinoid agonists increases the baseline firing rate (Cheer et al. 2000, French et al. 1997) and burst firing (Cheer et al. 2003, Diana et al. 1998, French et al. 1997) of VTA dopamine neurons and stimulates phasic dopamine release in the NAc (Cheer et al. 2004) in a CB1 receptor–dependent manner. However, dopamine neurons do not express CB1 receptors (Herkenham et al. 1991, Julian et al. 2003), and current data support a CB1 receptor–mediated increase in dopamine neuron activity due to local disinhibitory mechanisms (Lupica & Riegel 2005). During periods of burst firing, VTA dopamine neurons synthesize and release eCBs onto presynaptic GABAergic terminals expressing CB1 receptors, suppressing GABA-mediated inhibition and initiating a disinhibitory feedback loop that enhances dopamine cell firing (Alger & Kim 2011, Lupica & Riegel 2005, Melis et al. 2004, Riegel & Lupica 2004).

Although these actions on the mesolimbic dopamine system and associated abuse liability inherently limit the use of THC as a medicinal agent, its mechanism of action at the CB1 receptor reveals an opportunity to target the reward circuitry for therapeutic purposes. Indeed, CB1 receptor antagonism was investigated for a possible role in attenuating aberrant reward-seeking behavior. Preclinical animal work demonstrated significant reductions in abused drug–induced phasic dopamine release (Cheer et al. 2007), drug-seeking behavior (Cippitelli et al. 2005, Cohen et al. 2004, De Vries et al. 2001, Fattore et al. 2005, Justinova et al. 2008), and feeding behavior (Di Marzo & Matias 2005, Rinaldi-Carmona et al. 2004) due to functional antagonism of the CB1 receptor by the inverse agonist rimonabant. Owing to its effects on appetite and reward seeking, rimonabant was approved and marketed for the treatment of metabolic syndrome in obese patients (Christensen et al. 2007, Ward & Raffa 2011) and went to advanced clinical trials for smoking cessation (Cohen et al. 2004, Gelfand & Cannon 2006). However, even with psychiatric exclusions, clinical trials revealed adverse side effects of high doses, including anxiety, depression, and suicidal ideations (Christensen et al. 2007). Part of the failure of rimonabant is thought to be due to its action as an inverse agonist of the CB1 receptor, producing an effect intrinsically opposite to that of the classical agonist (Ward & Raffa 2011). Currently, neutral antagonists for the CB1 receptor are under investigation for their effects on food intake and reward seeking (see sidebar, Δ9-Tetrahydrocannabivarin) (Riedel et al. 2009, Tudge et al. 2015, Ward & Raffa 2011). Nevertheless, these results emphasize the critical involvement of the CB1 receptor in motivation for reward and highlight the need for alternative receptor targets for the treatment of motivational disorders.

Cannabidiol

Although the pCBs share structural similarities, they vary significantly in their receptor interactions. In contrast to THC, CBD does not target CB1 and CB2 receptors directly (McPartland et al. 2015), does not increase VTA dopamine cell firing (French et al. 1997), and does not have inherent rewarding properties (Katsidoni et al. 2013, Parker et al. 2004). Although early studies indicated that CBD was a CB1 receptor inverse agonist similar to rimonabant (Pertwee 2008, Thomas et al. 2007), a recent review of the literature does not support this conclusion (McPartland et al. 2015). Rather, accumulating evidence reveals that CBD has low binding affinity for the CB1 receptor (Bisogno et al. 2001, Jones et al. 2010, Pertwee 2008) and fails to elicit measurable activity in efficacy assays (Breivogel et al. 2001, Thomas et al. 2007), making CBD unlikely to convey the negative psychiatric side effects of rimonabant (McPartland et al. 2015). However, additional studies have demonstrated indirect CBD-mediated agonism and antagonism of the CB1 receptor (Table 1). Various in vivo effects of CBD are absent in CB1 receptor knockout mice (Wolf et al. 2010), suggesting that CBD may act as an indirect agonist. This conclusion is reinforced by data demonstrating CBD-induced increases in AEA levels due to inhibition of fatty acid amide hydrolase (FAAH), the hydrolytic enzyme of AEA (Bisogno et al. 2001, De Petrocellis et al. 2011), and inhibition of AEA uptake by the putative AEA transporter (Bisogno et al. 2001). Furthermore, CBD noncompetitively antagonizes CB1 receptor agonists in in vitro assays by an unknown mechanism (Thomas et al. 2007). Taken together, these findings suggest that although CBD is not a primary ligand of the CB1 receptor, it may influence CB1 receptor transmission by augmenting eCB tone or by other indirect methods.

Table 1.

Cannabidiol receptor targets with potential for modulating symptoms of motivational disorders

Receptor system Mechanism Potential therapeutic application References
eCB FAAH inhibitor Intestinal motility; augmenting eCB tone; addiction; depression Bisogno et al. 2001, Ligresti et al. 2006, Capasso et al. 2008, Leweke et al. 2012, Campos et al. 2013, Panlilio et al. 2013, Blanco et al. 2014, Chauvet et al. 2014
AEA uptake inhibitor Augmenting eCB tone; addiction; depression Rakhshan et al. 2000, Bisogno et al. 2001, Ligresti et al. 2006, Scherma et al. 2008, Gamaleddin et al. 2011, Leweke et al. 2012, Campos et al. 2013
Antagonist of CB1/CB2 receptor agonists Reduction of THC effects Thomas et al. 2007, Pertwee 2008
Expanded eCB GPR55 antagonist To be determined Pertwee 2007, Ryberg et al. 2007, Rahimi et al. 2015
TRPV1 agonist Schizophrenia; analgesia Costa et al. 2007, De Petrocellis et al. 2008, Santos et al. 2008, Terzian et al. 2009, Adamczyk et al. 2012, Heng et al. 2014
TRPV2 agonist Vasodilation Qin et al. 2008
TRPA1 agonist Analgesia De Petrocellis et al. 2008
TRPM8 antagonist Analgesia; prostate cancer De Petrocellis et al. 2008
Serotonin 5-HT1A agonist Addiction; anxiety; depression; schizophrenia Russo et al. 2005, Alex & Pehek 2007, Campos & Guimarães 2008, Resstel et al. 2009
5-HT2A agonist Addiction; schizophrenia Russo et al. 2005, Alex & Pehek 2007
5-HT3 agonist Addiction Alex & Pehek 2007, Xiong et al. 2012
Tryptophan degradation inhibitor Addiction; anxiety; depression; pain; inflammation Jenny et al. 2009, Ross 2009
Adenosine Adenosine uptake inhibitor and indirect A2A agonist Addiction; inflammation Carrier et al. 2006, Liou et al. 2008, Ferré et al. 2010
Opioid Mu-opioid ligand Addiction; pain Pertwee et al. 2002
Mu- and delta-opioid positive allosteric modulator Addiction; pain Kathmann et al. 2006
Dopamine D2 allosteric modulator Addiction Bloom & Hillard 1985
Dopamine transporter inhibitor Addiction; depression Pandolfo et al. 2011
Other GABAA positive allosteric modulator Addiction Long et al. 2012
Alpha-1 and alpha-1-beta glycine receptor positive allosteric modulator Pain; inflammation Ahrens et al. 2009
Regulator of intracellular calcium Epilepsy; neuroprotection Drysdale et al. 2006, Ryan et al. 2006
T-type calcium channel inhibitor Epilepsy; neuroprotection; pain Ross 2009
PPARγ agonist Vasorelaxation; addiction O’Sullivan et al. 2009, Enayatfard et al. 2013, Panlilio et al. 2013, de Guglielmo et al. 2014

Abbreviations: 5-HT, 5-hydroxytryptamine; AEA, N-arachidonoylethanolamine; CB1 and CB2, cannabinoid receptor type 1 and 2; eCB, endocannabinoid; FAAH, fatty acid amide hydrolase; PPARγ, peroxisome proliferator-activated receptor gamma; THC, Δ9-tetrahydrocannabinol; TRPA1, transient receptor potential cation channel A1; TRPM8, transient receptor potential cation channel M8; TRPV1 and TRPV2, transient receptor potential cation channel V1 and V2.

Apart from its effects on the eCB system, CBD exhibits promiscuous pharmacological activity across a range of receptor targets (Table 1). Several of these receptors may be considered part of an expanded eCB system owing to their affinity for various cannabinoid ligands (McPartland et al. 2015). In particular, the orphan GPCR GPR55 (Ross 2009) and the transient receptor potential (TRP) channels (Di Marzo et al. 2002) have been characterized as potential novel metabotropic and ionotropic cannabinoid receptors, respectively. In addition to actions within the expanded eCB system, CBD inhibits adenosine uptake (Table 1) and is both a direct and indirect 5-hydroxytryptamine 1A (5-HT1A) receptor agonist. It is also an allosteric modulator of mu- and delta-opioid, dopamine D2, GABAA, and glycine receptors. To date, most of the work on the molecular targets of CBD has been done only in vitro, and future studies will need to examine their therapeutic relevance in vivo.

THERAPEUTIC ACTIONS OF CANNABIDIOL IN DISORDERS OF MOTIVATION

Reward deficits and consequent motivational dysfunction are shared among affective disorders such as depression and anxiety and are an inherent feature of drug addiction (Russo & Nestler 2013). Comorbidity among these disorders is highly prevalent, with as many as 30–40% of individuals suffering from drug addiction also meeting criteria for depression or an anxiety disorder (Conway et al. 2006). This suggests a degree of potential overlap among the neurobiological mechanisms of these disorders, including disruptions of the mesolimbic reward circuitry and its interactions with other brain areas (Nestler & Carlezon 2006, Russo & Nestler 2013). As such, pharmacotherapies targeting these substrates may have beneficial outcomes across these disorders. Here we review the potential applications for CBD in the treatment of drug addiction, anxiety, and depression.

Drug Addiction

Drug addiction is a motivational disorder in which an individual compulsively seeks to use drugs and loses control over drug intake (Koob & Volkow 2009). Relapse to drug use after a period of withdrawal is one of the greatest challenges in the treatment of addiction, and among all the classes of abused drugs, there are currently few effective pharmacotherapies (Potenza et al. 2011). However, in preclinical studies in humans and animals, CBD reduces drug-motivated behavior (Morgan et al. 2013, Ren et al. 2009), attenuates withdrawal effects (Bhargava 1976; Chesher & Jackson 1985; Hine et al. 1975a, b), and limits cravings (Hurd et al. 2015). Consistent with results demonstrating antagonizing effects of CBD on THC-induced pharmacological actions, cannabis containing higher versus lower levels of CBD decreases the incentive salience of cannabis-related stimuli in smokers (Morgan et al. 2013), and a case study reported a reduction in cannabis withdrawal symptoms following CBD administration (Crippa et al. 2013).

Yet the majority of the treatment effects of CBD have been investigated in the context of opiate drugs. In morphine-dependent rats, CBD reduces withdrawal signs (Bhargava 1976; Chesher & Jackson 1985; Hine et al. 1975a,b) and enhances THC-meditated attenuation of abstinence scores (Hine et al. 1975a,b). Furthermore, although CBD does not reduce heroin self-administration or heroin-primed reinstatement of drug seeking in an animal model of relapse, it nonetheless diminishes heroin cue-primed drug seeking and normalizes heroin-induced impairments in accumbal AMPA and CB1 receptor levels (Ren et al. 2009). In an extension of these findings, a human clinical study demonstrated that CBD does not alter the subjective effects of fentanyl but attenuates heroin cue-induced drug craving and anxiety (Hurd et al. 2015). These results indicate that CBD effectively reduces opioid-paired cue reactivity but has little effect on the acute reinforcing properties of opioids. However, additional work shows that the reward-facilitating effects of morphine on intracranial self-stimulation (ICSS) are decreased by systemic CBD administration, and these effects are blocked by injection of the selective 5-HT1A receptor antagonist WAY-100635 into the dorsal raphe (Katsidoni et al. 2013). This finding implicates dorsal raphe 5-HT1A autoreceptors in the mitigating effects of CBD on morphine reward. Importantly, CBD is an agonist of the 5-HT1A receptor (Table 1), and evidence suggests that infusion of 5-HT1A agonists directly into the dorsal raphe reduces baseline serotonin and dopamine release in the NAc (Yoshimoto & McBride 1992), suggesting a mechanism whereby CBD may reduce the acute reinforcing effects of morphine. However, the 5-HT1A receptor is also localized postsynaptically in dopamine terminal regions such as the prefrontal cortex, amygdala, and hippocampus as well as the NAc (Pompeiano et al. 1992). Therefore, future investigations concentrating on the actions of CBD on 5-HT1A receptors within the dorsal raphe and elsewhere in the brain reward circuitry may yield promising treatment outcomes for opioid addiction.

In contrast to its effects on opioid-motivated behaviors, CBD has less apparent influence on psychostimulant reward and reinforcement. Administration of CBD fails to alter cocaine-mediated decreases in ICSS thresholds (Katsidoni et al. 2013) or disrupt cocaine- and amphetamine-conditioned place preference (Parker et al. 2004). An exception to this lack of therapeutic effect relates to nicotine reinforcement. Preliminary findings from a pilot study in humans revealed that CBD reduced cigarette smoking significantly compared to placebo in smokers trying to quit (Morgan et al. 2013). Although the mechanism for this effect has not been investigated explicitly, CBD may modulate nicotine reward through its ability to increase eCB tone by FAAH inhibition (Table 1). Other work has demonstrated that inhibiting FAAH prevents nicotine seeking (Forget et al. 2009, Scherma et al. 2008), nicotine-induced dopamine release in the NAc (Scherma et al. 2008), and anxiety during nicotine withdrawal (Cippitelli et al. 2011) in animals. These results suggest that, although the therapeutic effects of CBD on nicotine reward are promising, additional work is crucially needed to outline its precise mechanism of action.

Anxiety

Clinical studies have revealed robust anxiolytic effects of CBD. For example, CBD reversed anxiety generated by a high dose of THC (Zuardi et al. 1982) and by a public-speaking simulation in both patients with social phobia (Bergamaschi et al. 2011) and controls (Crippa et al. 2004, Zuardi et al. 1993). Neuroimaging studies also showed that CBD decreased activation of brain regions associated with anxiety, fear, and emotional processing, including the amygdala and the anterior and posterior cingulate cortex (Crippa et al. 2004, Fusar-Poli et al. 2010).

In rodents, the anxiolytic effects of CBD are also well established across numerous different behavioral paradigms. Results have indicated a clear inverted U-shaped dose-response curve for the effects of CBD on anxiety measures, with lower doses more efficacious than higher doses (Guimarães et al. 1990). Anxiolytic properties of systemic CBD have been confirmed in the elevated plus maze (Campos & Guimarães 2008, Gomes et al. 2010, Guimarães et al. 1990, Onaivi et al. 1990), elevated T maze (de Paula Soares et al. 2010), Vogel conflict test (Campos & Guimarães 2008, Gomes et al. 2010, Moreira et al. 2006), and contextual fear conditioning model (Resstel et al. 2006). Moreover, direct microinjection of CBD into several brain regions identified sites involved in its antianxiety effects. CBD administration into the central nucleus of the amygdala (Hsiao et al. 2012), bed nucleus of the stria terminalis (BNST) (Gomes et al. 2010, 2012), and dorsal periaqueductal gray (DPAG) (Campos & Guimarães 2008, de Paula Soares et al. 2010) promotes anxiolytic-like effects in behavioral anxiety procedures; within the BNST (Gomes et al. 2010, 2012) and DPAG (Campos & Guimarães 2008, de Paula Soares et al. 2010), these effects are blocked by preadministration of a 5-HT1A receptor antagonist. Together, these findings strongly implicate 5-HT-mediated transmission in key brain areas as a potential mechanism underlying the effects of CBD on anxiety.

Depression

Stress is a predisposing factor for anxiety and depression (Kessler 1997), and many preclinical tests of depression involve acute or chronic stress and inescapable aversive stimuli and often induce both depression- and anxiety-like symptoms (Campos et al. 2012, Russo & Nestler 2013). In such tests, CBD reduces autonomic indices of stress (Resstel et al. 2009) and behavioral manifestations of depression and anxiety (El-Alfy et al. 2010, Granjeiro et al. 2011, Resstel et al. 2009, Zanelati et al. 2010). For example, increases in heart rate and blood pressure during restraint stress as well as anxiety-like performance on an elevated plus maze following restraint stress are mitigated by systemically (Resstel et al. 2009) and centrally (Granjeiro et al. 2011) administered CBD. Additionally, although CBD does not modify the tail suspension test (El-Alfy et al. 2010), it significantly reduces immobility in the forced swim test (El-Alfy et al. 2010, Zanelati et al. 2010). On these tasks, the antidepressant and anxiolytic effects of CBD are likely 5-HT1A-mediated, as they are blocked by the 5-HT1A receptor antagonist WAY-100635 (Resstel et al. 2009, Zanelati et al. 2010).

However, similar to classical antidepressants, CBD may also reduce depressive symptoms by stimulating hippocampal neurogenesis (Campos et al. 2013, David et al. 2010). This is supported by recent work demonstrating that repeated administration of CBD in mice exposed to chronic unpredictable stress reduced depressive- and anxiety-like behaviors and stimulated hippocampal progenitor proliferation and neurogenesis. Moreover, these beneficial actions of CBD are prevented by genetic ablation of proliferating progenitor cells (Campos et al. 2013). The mechanism whereby CBD stimulates neurogenesis is hypothesized to depend on elevation of hippocampal AEA levels, as overexpression of FAAH inhibits CBD-induced cell proliferation. Interestingly, a clinical study found that whereas serum levels of AEA were negatively correlated with anxiety in patients with major depression, they were not associated with major depressive symptoms (Hill et al. 2008). However, in patients with minor depression, AEA levels were increased compared to control levels, suggesting a neuroprotective or compensatory mechanism in patients with less severe depressive symptoms. The significance of AEA levels and eCB tone and their moderation by CBD in depression warrants further research. Nevertheless, present results signify an important therapeutic role for CBD in the treatment of depression and anxiety.

CONCLUSIONS AND FUTURE DIRECTIONS

Results from a growing number of preclinical and clinical studies identify a novel treatment application for CBD in disorders of motivation, including drug addiction, anxiety, and depression (Table 1). A review of the findings suggests that CBD may attenuate motivational dysfunction through activation of the 5-HT1A receptor and elevations in eCB tone. Given the multitude of molecular targets for CBD, there is substantial potential for additional beneficial effects through actions at other receptors. Further in vivo exploration of these targets will be fundamental for developing a thorough understanding of the therapeutic efficacy of CBD for drug addiction and affective disorders. Critically, isolating the mechanisms of CBD may pinpoint selective targets for rational drug development. Yet the greatest treatment value of CBD may lie in its multitarget actions or polypharmacology (Hopkins 2008). Pharmacotherapies that target numerous receptors across neural networks may be more efficacious than those that are maximally selective for a single target (Hopkins 2008, Mencher & Wang 2005). Evidence for this comes from the use of antidepressants and antipsychotics, which derive their therapeutic effects via interactions across various GPCRs (Anighoro et al. 2014). Therefore, in motivational disorders with complex etiology and underlying neural substrates, the multitarget effects of CBD may make it a highly efficacious treatment option.

Δ9-TETRAHYDROCANNABIVARIN.

In addition to CBD, other pCBs may exhibit novel therapeutic effects in the treatment of psychiatric disorders of motivation. Of particular interest is Δ9-tetrahydrocannabivarin (THCV). THCV is a pCB with multitarget pharmacological effects as a neutral antagonist of the CB1 receptor as well as a partial agonist of the CB2 receptor (McPartland et al. 2015). Neutral antagonism of the CB1 receptor may allow THCV to therapeutically target the mesolimbic reward circuitry and avoid the negative psychiatric side effects of CB1 receptor inverse agonists such as rimonabant (Riedel et al. 2009, Ward & Raffa 2011, Tudge et al. 2015). Similarly, the actions of THCV as a CB2 receptor partial agonist may engender it with additional therapeutic potential for dysregulated reward-motivated behaviors. Recent work revealed the expression of CB2 receptors in VTA dopamine neurons (Zhang et al. 2014), and activation of these receptors reduced dopamine firing and cocaine self-administration (Xi et al. 2011, Zhang et al. 2014). Given these promising results, additional research on the medicinal value of pCBs such as THCV in disorders of reward and motivation is warranted.

Acknowledgments

The authors would like to thank Drs. Dan Covey and Jennifer Wenzel for helpful comments on the manuscript. This work was supported by National Institute on Drug Abuse grant DA022340 to J.F.C.

Glossary

pCB

phytocannabinoid

THC

Δ9-tetrahydrocannabinol

CBD

cannabidiol

eCB

endocannabinoid

GPCR

G protein–coupled receptor

CB1 receptor

cannabinoid type 1 receptor

CB2 receptor

cannabinoid type 2 receptor

AEA

N-arachidonoylethanolamine or anandamide

NAc

nucleus accumbens

VTA

ventral tegmental area

GABA

gamma-aminobutyric acid

FAAH

fatty acid amide hydrolase

TRP

transient receptor potential

5-HT

5-hydroxytryptamine or serotonin

BNST

bed nucleus of the stria terminalis

DPAG

dorsal periaqueductal gray

Footnotes

DISCLOSURE STATEMENT

The authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review.

LITERATURE CITED

  1. Adamczyk P, Miszkiel J, McCreary AC, Filip M, Papp M, Przegaliński E. The effects of cannabinoid CB1, CB2 and vanilloid TRPV1 receptor antagonists on cocaine addictive behavior in rats. Brain Res. 2012;1444:45–54. doi: 10.1016/j.brainres.2012.01.030. [DOI] [PubMed] [Google Scholar]
  2. Ahrens J, Demir R, Leuwer M, de la Roche J, Krampfl K, et al. The nonpsychotropic cannabinoid cannabidiol modulates and directly activates alpha-1 and alpha-1-beta glycine receptor function. Pharmacology. 2009;83(4):217–22. doi: 10.1159/000201556. [DOI] [PubMed] [Google Scholar]
  3. Alex KD, Pehek EA. Pharmacologic mechanisms of serotonergic regulation of dopamine neurotransmission. Pharmacol Ther. 2007;113(2):296–320. doi: 10.1016/j.pharmthera.2006.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alger BE. Endocannabinoid signaling in neural plasticity. Curr Top Behav Neurosci. 2009;1:141–72. doi: 10.1007/978-3-540-88955-7_6. [DOI] [PubMed] [Google Scholar]
  5. Alger BE, Kim J. Supply and demand for endocannabinoids. Trends Neurosci. 2011;34(6):304–15. doi: 10.1016/j.tins.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Anighoro A, Bajorath J, Rastelli G. Polypharmacology: challenges and opportunities in drug discovery. J Med Chem. 2014;57(19):7874–87. doi: 10.1021/jm5006463. [DOI] [PubMed] [Google Scholar]
  7. Barnes MP. Sativex: clinical efficacy and tolerability in the treatment of symptoms of multiple sclerosis and neuropathic pain. Expert Opin Pharmacother. 2006;7(5):607–15. doi: 10.1517/14656566.7.5.607. [DOI] [PubMed] [Google Scholar]
  8. Ben-Shabat S, Fride E, Sheskin T, Tamiri T, Rhee MH, et al. An entourage effect: Inactive endogenous fatty acid glycerol esters enhance 2-arachidonoyl-glycerol cannabinoid activity. Eur J Pharmacol. 1998;353(1):23–31. doi: 10.1016/s0014-2999(98)00392-6. [DOI] [PubMed] [Google Scholar]
  9. Bergamaschi MM, Queiroz RHC, Chagas MHN, de Oliveira DCG, De Martinis BS, et al. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naïve social phobia patients. Neuropsychopharmacology. 2011;36(6):1219–26. doi: 10.1038/npp.2011.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bhargava HN. Effect of some cannabinoids on naloxone-precipitated abstinence in morphine-dependent mice. Psychopharmacology. 1976;49(3):267–70. doi: 10.1007/BF00426828. [DOI] [PubMed] [Google Scholar]
  11. Bidaut-Russell M, Devane WA, Howlett AC. Cannabinoid receptors and modulation of cyclic AMP accumulation in the rat brain. J Neurochem. 1990;55(1):21–26. doi: 10.1111/j.1471-4159.1990.tb08815.x. [DOI] [PubMed] [Google Scholar]
  12. Bisogno T, Hanuš L, De Petrocellis L, Tchilibon S, Ponde DE, et al. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br J Pharmacol. 2001;134(4):845–52. doi: 10.1038/sj.bjp.0704327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Blanco E, Pavón FJ, Palomino A, Luque-Rojas MJ, Serrano A, et al. Cocaine-induced behavioral sensitization is associated with changes in the expression of endocannabinoid and glutamatergic signaling systems in the mouse prefrontal cortex. Int J Neuropsychopharmacol. 2014;18(1):pyu024. doi: 10.1093/ijnp/pyu024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bloom AS, Hillard CJ. Cannabinoids, neurotransmitter receptors and brain membranes. In: Harvey DJ, editor. Marihuana ’84. Oxford, UK: IRL; 1985. pp. 217–31. [Google Scholar]
  15. Breivogel CS, Griffin G, Di Marzo V, Martin BR. Evidence for a new G protein-coupled cannabinoid receptor in mouse brain. Mol Pharmacol. 2001;60(1):155–63. [PubMed] [Google Scholar]
  16. Campos AC, Guimarães FS. Involvement of 5HT1A receptors in the anxiolytic-like effects of cannabidiol injected into the dorsolateral periaqueductal gray of rats. Psychopharmacology. 2008;199(2):223–30. doi: 10.1007/s00213-008-1168-x. [DOI] [PubMed] [Google Scholar]
  17. Campos AC, Moreira FA, Gomes FV, Del Bel EA, Guimarães FS. Multiple mechanisms involved in the large-spectrum therapeutic potential of cannabidiol in psychiatric disorders. Philos Trans R Soc B Biol Sci. 2012;367(1607):3364–78. doi: 10.1098/rstb.2011.0389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Campos AC, Ortega Z, Palazuelos J, Fogaça MV, Aguiar DC, et al. The anxiolytic effect of cannabidiol on chronically stressed mice depends on hippocampal neurogenesis: involvement of the endocannabinoid system. Int J Neuropsychopharmacol. 2013;16(6):1407–19. doi: 10.1017/S1461145712001502. [DOI] [PubMed] [Google Scholar]
  19. Capasso R, Borrelli F, Aviello G, Romano B, Scalisi C, et al. Cannabidiol, extracted from Cannabis sativa, selectively inhibits inflammatory hypermotility in mice. Br J Pharmacol. 2008;154(5):1001–8. doi: 10.1038/bjp.2008.177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Carrier EJ, Auchampach JA, Hillard CJ. Inhibition of an equilibrative nucleoside transporter by cannabidiol: a mechanism of cannabinoid immunosuppression. PNAS. 2006;103(20):7895–900. doi: 10.1073/pnas.0511232103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Chauvet C, Nicolas C, Thiriet N, Lardeux MV, Duranti A, Solinas M. Chronic stimulation of the tone of endogenous anandamide reduces cue- and stress-induced relapse in rats. Int J Neuropsychopharmacol. 2014;18(1):pyu025. doi: 10.1093/ijnp/pyu025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Cheer JF, Kendall DA, Mason R, Marsden CA. Differential cannabinoid-induced electrophysiological effects in rat ventral tegmentum. Neuropharmacology. 2003;44(5):633–41. doi: 10.1016/s0028-3908(03)00029-7. [DOI] [PubMed] [Google Scholar]
  23. Cheer JF, Marsden CA, Kendall DA, Mason R. Lack of response suppression follows repeated ventral tegmental cannabinoid administration: an in vitro electrophysiological study. Neuroscience. 2000;99(4):661–67. doi: 10.1016/s0306-4522(00)00241-4. [DOI] [PubMed] [Google Scholar]
  24. Cheer JF, Wassum KM, Heien MLAV, Phillips PEM, Wightman RM. Cannabinoids enhance subsecond dopamine release in the nucleus accumbens of awake rats. J Neurosci. 2004;24(18):4393–400. doi: 10.1523/JNEUROSCI.0529-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Cheer JF, Wassum KM, Sombers LA, Heien MLAV, Ariansen JL, et al. Phasic dopamine release evoked by abused substances requires cannabinoid receptor activation. J Neurosci. 2007;27(4):791–95. doi: 10.1523/JNEUROSCI.4152-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Chesher GB, Jackson DM. The quasi-morphine withdrawal syndrome: effect of cannabinol, cannabidiol and tetrahydrocannabinol. Pharmacol Biochem Behav. 1985;23(1):13–15. doi: 10.1016/0091-3057(85)90122-4. [DOI] [PubMed] [Google Scholar]
  27. Christensen R, Kristensen PK, Bartels EM, Bliddal H, Astrup A. Efficacy and safety of the weight-loss drug rimonabant: a meta-analysis of randomised trials. Lancet. 2007;370(9600):1706–13. doi: 10.1016/S0140-6736(07)61721-8. [DOI] [PubMed] [Google Scholar]
  28. Cilio MR, Thiele EA, Devinsky O. The case for assessing cannabidiol in epilepsy. Epilepsia. 2014;55(6):787–90. doi: 10.1111/epi.12635. [DOI] [PubMed] [Google Scholar]
  29. Cippitelli A, Astarita G, Duranti A, Caprioli G, Ubaldi M, et al. Endocannabinoid regulation of acute and protracted nicotine withdrawal: effect of FAAH inhibition. PLOS ONE. 2011;6(11):e28142. doi: 10.1371/journal.pone.0028142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Cippitelli A, Bilbao A, Hansson AC, Del Arco I, Sommer W, et al. Cannabinoid CB1 receptor antagonism reduces conditioned reinstatement of ethanol-seeking behavior in rats. Eur J Neurosci. 2005;21(8):2243–51. doi: 10.1111/j.1460-9568.2005.04056.x. [DOI] [PubMed] [Google Scholar]
  31. Cohen C, Perrault G, Griebel G, Soubrié P. Nicotine-associated cues maintain nicotine-seeking behavior in rats several weeks after nicotine withdrawal: reversal by the cannabinoid (CB1) receptor antagonist, rimonabant (SR141716) Neuropsychopharmacology. 2004;30(1):145–55. doi: 10.1038/sj.npp.1300541. [DOI] [PubMed] [Google Scholar]
  32. Conway KP, Compton W, Stinson FS, Grant BF. Lifetime comorbidity of DSM-IV mood and anxiety disorders and specific drug use disorders: results from the National Epidemiologic Survey on Alcohol and Related Conditions. J Clin Psychiatry. 2006;67(2):247–57. doi: 10.4088/jcp.v67n0211. [DOI] [PubMed] [Google Scholar]
  33. Costa B, Trovato AE, Comelli F, Giagnoni G, Colleoni M. The non-psychoactive cannabis constituent cannabidiol is an orally effective therapeutic agent in rat chronic inflammatory and neuropathic pain. Eur J Pharmacol. 2007;556(1–3):75–83. doi: 10.1016/j.ejphar.2006.11.006. [DOI] [PubMed] [Google Scholar]
  34. Crippa JAS, Zuardi AW, Garrido GEJ, Wichert-Ana L, Guarnieri R, et al. Effects of cannabidiol (CBD) on regional cerebral blood flow. Neuropsychopharmacology. 2004;29(2):417–26. doi: 10.1038/sj.npp.1300340. [DOI] [PubMed] [Google Scholar]
  35. Crippa JAS, Hallak JEC, Machado-de-Sousa JP, Queiroz RHC, Bergamaschi M, et al. Cannabidiol for the treatment of cannabis withdrawal syndrome: a case report. J Clin Pharm Ther. 2013;38(2):162–64. doi: 10.1111/jcpt.12018. [DOI] [PubMed] [Google Scholar]
  36. David DJ, Wang J, Samuels BA, Rainer Q, David I, et al. Implications of the functional integration of adult-born hippocampal neurons in anxiety-depression disorders. Neuroscientist. 2010;16(5):578–91. doi: 10.1177/1073858409360281. [DOI] [PubMed] [Google Scholar]
  37. de Guglielmo G, Kallupi M, Scuppa G, Stopponi S, Demopulos G, et al. Analgesic tolerance to morphine is regulated by PPARγ. Br J Pharmacol. 2014;171(23):5407–16. doi: 10.1111/bph.12851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. de Meijer EPM, Bagatta M, Carboni A, Crucitti P, Moliterni VMC, et al. The inheritance of chemical phenotype in Cannabis sativa L. Genetics. 2003;163(1):335–46. doi: 10.1093/genetics/163.1.335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. de Paula Soares V, Campos AC, Camin de Bortoli V, Zangrossi H, Jr, Guimarães FS, Zuardi AW. Intra-dorsal periaqueductal gray administration of cannabidiol blocks panic-like response by activating 5-HT1A receptors. Behav Brain Res. 2010;213(2):225–29. doi: 10.1016/j.bbr.2010.05.004. [DOI] [PubMed] [Google Scholar]
  40. De Petrocellis L, Ligresti A, Moriello AS, Allarà M, Bisogno T, et al. Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br J Pharmacol. 2011;163(7):1479–94. doi: 10.1111/j.1476-5381.2010.01166.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. De Petrocellis L, Vellani V, Schiano-Moriello A, Marini P, Magherini PC, et al. Plant-derived cannabinoids modulate the activity of transient receptor potential channels of ankyrin type-1 and melastatin type-8. J Pharmacol Exp Ther. 2008;325(3):1007–15. doi: 10.1124/jpet.107.134809. [DOI] [PubMed] [Google Scholar]
  42. Devane WA, Dysarz FA, Johnson MR, Melvin LS, Howlett AC. Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol. 1988;34(5):605–13. [PubMed] [Google Scholar]
  43. Devane WA, Hanuš L, Breuer A, Pertwee RG, Stevenson LA, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992;258(5090):1946–49. doi: 10.1126/science.1470919. [DOI] [PubMed] [Google Scholar]
  44. Devinsky O, Cilio MR, Cross H, Fernandez-Ruiz J, French J, et al. Cannabidiol: pharmacology and potential therapeutic role in epilepsy and other neuropsychiatric disorders. Epilepsia. 2014;55(6):791–802. doi: 10.1111/epi.12631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. De Vries TJ, Shaham Y, Homberg JR, Crombag H, Schuurman K, et al. A cannabinoid mechanism in relapse to cocaine seeking. Nat Med. 2001;7(10):1151–54. doi: 10.1038/nm1001-1151. [DOI] [PubMed] [Google Scholar]
  46. Diana M, Melis M, Gessa GL. Increase in mesoprefrontal dopaminergic activity after stimulation of CB1 receptors by cannabinoids. Eur J Neurosci. 1998;10(9):2825–30. doi: 10.1111/j.1460-9568.1998.00292.x. [DOI] [PubMed] [Google Scholar]
  47. Di Chiara G, Imperato A. Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. PNAS. 1988;85(14):5274–78. doi: 10.1073/pnas.85.14.5274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Di Marzo V, De Petrocellis L, Fezza F, Ligresti A, Bisogno T. Anandamide receptors. Prostaglandins Leukot Essent Fat Acids. 2002;66(2–3):377–91. doi: 10.1054/plef.2001.0349. [DOI] [PubMed] [Google Scholar]
  49. Di Marzo V, Matias I. Endocannabinoid control of food intake and energy balance. Nat Neurosci. 2005;8(5):585–89. doi: 10.1038/nn1457. [DOI] [PubMed] [Google Scholar]
  50. Drysdale AJ, Ryan D, Pertwee RG, Platt B. Cannabidiol-induced intracellular Ca2+ elevations in hippocampal cells. Neuropharmacology. 2006;50(5):621–31. doi: 10.1016/j.neuropharm.2005.11.008. [DOI] [PubMed] [Google Scholar]
  51. El-Alfy AT, Ivey K, Robinson K, Ahmed S, Radwan M, et al. Antidepressant-like effect of Δ9-tetrahydrocannabinol and other cannabinoids isolated from Cannabis sativa L. Pharmacol Biochem Behav. 2010;95(4):434–42. doi: 10.1016/j.pbb.2010.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Enayatfard L, Rostami F, Nasoohi S, Oryan S, Ahmadiani A, Dargahi L. Dual role of PPAR-γ in induction and expression of behavioral sensitization to cannabinoid receptor agonist WIN55,212–2. Neuromolecular Med. 2013;15(3):523–35. doi: 10.1007/s12017-013-8238-x. [DOI] [PubMed] [Google Scholar]
  53. Fattore L, Spano S, Cossu G, Deiana S, Fadda P, Fratta W. Cannabinoid CB1 antagonist SR 141716A attenuates reinstatement of heroin self-administration in heroin-abstinent rats. Neuropharmacology. 2005;48(8):1097–104. doi: 10.1016/j.neuropharm.2005.01.022. [DOI] [PubMed] [Google Scholar]
  54. Fankhauser M. History of cannabis in Western Medicine. In: Grotenhermen F, Russo E, editors. Cannabis and Cannabinoids. New York: Haworth Integr. Heal; 2002. pp. 37–51. [Google Scholar]
  55. Ferré S, Lluís C, Justinova Z, Quiroz C, Orru M, et al. Adenosine-cannabinoid receptor interactions: implications for striatal function. Br J Pharmacol. 2010;160(3):443–53. doi: 10.1111/j.1476-5381.2010.00723.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Forget B, Coen KM, Le Foll B. Inhibition of fatty acid amide hydrolase reduces reinstatement of nicotine seeking but not break point for nicotine self-administration—comparison with CB1 receptor blockade. Psychopharmacology. 2009;205(4):613–24. doi: 10.1007/s00213-009-1569-5. [DOI] [PubMed] [Google Scholar]
  57. French ED, Dillon K, Wu X. Cannabinoids excite dopamine neurons in the ventral tegmentum and substantia nigra. NeuroReport. 1997;8(3):649–52. doi: 10.1097/00001756-199702100-00014. [DOI] [PubMed] [Google Scholar]
  58. Freund TF, Katona I, Piomelli D. Role of endogenous cannabinoids in synaptic signaling. Physiol Rev. 2003;83(3):1017–66. doi: 10.1152/physrev.00004.2003. [DOI] [PubMed] [Google Scholar]
  59. Fusar-Poli P, Allen P, Bhattacharyya S, Crippa JAS, Mechelli A, et al. Modulation of effective connectivity during emotional processing by Δ9-tetrahydrocannabinol and cannabidiol. Int J Neuropsychopharmacol. 2010;13(4):421–32. doi: 10.1017/S1461145709990617. [DOI] [PubMed] [Google Scholar]
  60. Gamaleddin I, Guranda M, Goldberg SR, Le Foll B. The selective anandamide transport inhibitor VDM11 attenuates reinstatement of nicotine seeking behaviour, but does not affect nicotine intake. Br J Pharmacol. 2011;164(6):1652–60. doi: 10.1111/j.1476-5381.2011.01440.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Gaoni Y, Mechoulam R. Isolation, structure, and partial synthesis of an active constituent of hashish. J Am Chem Soc. 1964;86(8):1646–47. [Google Scholar]
  62. Gelfand EV, Cannon CP. Rimonabant: a selective blocker of the cannabinoid CB1 receptors for the management of obesity, smoking cessation and cardiometabolic risk factors. Expert Opin Investig Drugs. 2006;15(3):307–15. doi: 10.1517/13543784.15.3.307. [DOI] [PubMed] [Google Scholar]
  63. Gomes FV, Reis DG, Alves FHF, Corrêa FMA, Guimarães FS, Resstel LBM. Cannabidiol injected into the bed nucleus of the stria terminalis reduces the expression of contextual fear conditioning via 5-HT1A receptors. J Psychopharmacol. 2012;26(1):104–13. doi: 10.1177/0269881110389095. [DOI] [PubMed] [Google Scholar]
  64. Gomes FV, Resstel LBM, Guimarães FS. The anxiolytic-like effects of cannabidiol injected into the bed nucleus of the stria terminalis are mediated by 5-HT1A receptors. Psychopharmacology. 2010;213(2–3):465–73. doi: 10.1007/s00213-010-2036-z. [DOI] [PubMed] [Google Scholar]
  65. Granjeiro ÉM, Gomes FV, Guimarães FS, Corrêa FMA, Resstel LBM. Effects of intracisternal administration of cannabidiol on the cardiovascular and behavioral responses to acute restraint stress. Pharmacol Biochem Behav. 2011;99(4):743–48. doi: 10.1016/j.pbb.2011.06.027. [DOI] [PubMed] [Google Scholar]
  66. Grinspoon L, Bakalar JB. Marijuana as medicine: a plea for reconsideration. JAMA. 1995;273(23):1875–76. [PubMed] [Google Scholar]
  67. Guimarães FS, Chiaretti TM, Graeff FG, Zuardi AW. Antianxiety effect of cannabidiol in the elevated plus-maze. Psychopharmacology. 1990;100(4):558–59. doi: 10.1007/BF02244012. [DOI] [PubMed] [Google Scholar]
  68. Heng L-J, Huang B, Guo H, Ma L-T, Yuan W-X, et al. Blocking TRPV1 in nucleus accumbens inhibits persistent morphine conditioned place preference expression in rats. PLOS ONE. 2014;9(8):e104546. doi: 10.1371/journal.pone.0104546. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  69. Henry DJ, Chavkin C. Activation of inwardly rectifying potassium channels (GIRK1) by co-expressed rat brain cannabinoid receptors in Xenopus oocytes. Neurosci Lett. 1995;186(2–3):91–94. doi: 10.1016/0304-3940(95)11289-9. [DOI] [PubMed] [Google Scholar]
  70. Herkenham M, Lynn AB, Johnson MR, Melvin LS, de Costa BR, Rice KC. Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci. 1991;11(2):563–83. doi: 10.1523/JNEUROSCI.11-02-00563.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Herkenham M, Lynn AB, Little MD, Johnson MR, Melvin LS, et al. Cannabinoid receptor localization in brain. PNAS. 1990;87(5):1932–36. doi: 10.1073/pnas.87.5.1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Hill AJ, Williams CM, Whalley BJ, Stephens GJ. Phytocannabinoids as novel therapeutic agents in CNS disorders. Pharmacol Ther. 2012;133(1):79–97. doi: 10.1016/j.pharmthera.2011.09.002. [DOI] [PubMed] [Google Scholar]
  73. Hill M, Miller G, Ho W-S, Gorzalka B, Hillard C. Serum endocannabinoid content is altered in females with depressive disorders: a preliminary report. Pharmacopsychiatry. 2008;41(2):48–53. doi: 10.1055/s-2007-993211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Hine B, Torrelio M, Gershon S. Differential effect of cannabinol and cannabidiol on THC-induced responses during abstinence in morphine-dependent rats. Res Commun Chem Pathol Pharmacol. 1975a;12(1):185–88. [PubMed] [Google Scholar]
  75. Hine B, Torrelio M, Gershon S. Interactions between cannabidiol and Δ9-THC during abstinence in morphine-dependent rats. Life Sci. 1975b;17(6):851–57. doi: 10.1016/0024-3205(75)90435-x. [DOI] [PubMed] [Google Scholar]
  76. Hoffman AF, Lupica CR. Mechanisms of cannabinoid inhibition of GABAA synaptic transmission in the hippocampus. J Neurosci. 2000;20(7):2470–79. doi: 10.1523/JNEUROSCI.20-07-02470.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Hopkins AL. Network pharmacology: the next paradigm in drug discovery. Nat Chem Biol. 2008;4(11):682–90. doi: 10.1038/nchembio.118. [DOI] [PubMed] [Google Scholar]
  78. Hsiao Y-T, Yi P-L, Li C-L, Chang F-C. Effect of cannabidiol on sleep disruption induced by the repeated combination tests consisting of open field and elevated plus-maze in rats. Neuropharmacology. 2012;62(1):373–84. doi: 10.1016/j.neuropharm.2011.08.013. [DOI] [PubMed] [Google Scholar]
  79. Huestis MA, Gorelick DA, Heishman SJ, Preston KL, Nelson RA, et al. Blockade of effects of smoked marijuana by the CB1-selective cannabinoid receptor antagonist SR141716. Arch Gen Psychiatry. 2001;58(4):322–28. doi: 10.1001/archpsyc.58.4.322. [DOI] [PubMed] [Google Scholar]
  80. Hurd YL, Yoon M, Manini AF, Hernandez S, Olmedo R, et al. Early phase in the development of cannabidiol as a treatment for addiction: opioid relapse takes initial center stage. Neurotherapeutics. 2015;12(4):807–15. doi: 10.1007/s13311-015-0373-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Jenny M, Santer E, Pirich E, Schennach H, Fuchs D. Δ9-Tetrahydrocannabinol and cannabidiol modulate mitogen-induced tryptophan degradation and neopterin formation in peripheral blood mononuclear cells in vitro. J Neuroimmunol. 2009;207(1–2):75–82. doi: 10.1016/j.jneuroim.2008.12.004. [DOI] [PubMed] [Google Scholar]
  82. Jones NA, Hill AJ, Smith I, Bevan SA, Williams CM, et al. Cannabidiol displays antiepileptiform and antiseizure properties in vitro and in vivo. J Pharmacol Exp Ther. 2010;332(2):569–77. doi: 10.1124/jpet.109.159145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Julian MD, Martin AB, Cuellar B, Rodriguez De Fonseca F, Navarro M, et al. Neuroanatomical relationship between type 1 cannabinoid receptors and dopaminergic systems in the rat basal ganglia. Neuroscience. 2003;119(1):309–18. doi: 10.1016/s0306-4522(03)00070-8. [DOI] [PubMed] [Google Scholar]
  84. Justinova Z, Munzar P, Panlilio LV, Yasar S, Redhi GH, et al. Blockade of THC-seeking behavior and relapse in monkeys by the cannabinoid CB1-receptor antagonist rimonabant. Neuropsychopharmacology. 2008;33(12):2870–77. doi: 10.1038/npp.2008.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Kathmann M, Flau K, Redmer A, Tränkle C, Schlicker E. Cannabidiol is an allosteric modulator at mu- and delta-opioid receptors. Naunyn-Schmiedeberg’s Arch Pharmacol. 2006;372(5):354–61. doi: 10.1007/s00210-006-0033-x. [DOI] [PubMed] [Google Scholar]
  86. Katsidoni V, Anagnostou I, Panagis G. Cannabidiol inhibits the reward-facilitating effect of morphine: involvement of 5-HT1A receptors in the dorsal raphe nucleus. Addict Biol. 2013;18(2):286–96. doi: 10.1111/j.1369-1600.2012.00483.x. [DOI] [PubMed] [Google Scholar]
  87. Kessler RC. The effects of stressful life events on depression. Annu Rev Psychol. 1997;48:191–214. doi: 10.1146/annurev.psych.48.1.191. [DOI] [PubMed] [Google Scholar]
  88. Koob GF, Volkow ND. Neurocircuitry of addiction. Neuropsychopharmacology. 2009;35(1):217–38. doi: 10.1038/npp.2009.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Leweke FM, Piomelli D, Pahlisch F, Muhl D, Gerth CW, et al. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl Psychiatry. 2012;2:e94. doi: 10.1038/tp.2012.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Li H-L. Hallucinogenic plants in Chinese herbals. J Psychoact Drugs. 1978;10(1):17–26. [Google Scholar]
  91. Ligresti A, Moriello AS, Starowicz K, Matias I, Pisanti S, et al. Antitumor activity of plant cannabinoids with emphasis on the effect of cannabidiol on human breast carcinoma. J Pharmacol Exp Ther. 2006;318(3):1375–87. doi: 10.1124/jpet.106.105247. [DOI] [PubMed] [Google Scholar]
  92. Liou GI, Auchampach JA, Hillard CJ, Zhu G, Yousufzai B, et al. Mediation of cannabidiol anti-inflammation in the retina by equilibrative nucleoside transporter and A2A adenosine receptor. Investig Ophthalmol Vis Sci. 2008;49(12):5526–31. doi: 10.1167/iovs.08-2196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Long LE, Chesworth R, Huang X-F, Wong A, Spiro A, et al. Distinct neurobehavioural effects of cannabidiol in transmembrane domain neuregulin 1 mutant mice. PLOS ONE. 2012;7(4):e34129. doi: 10.1371/journal.pone.0034129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Lupica CR, Riegel AC. Endocannabinoid release from midbrain dopamine neurons: a potential substrate for cannabinoid receptor antagonist treatment of addiction. Neuropharmacology. 2005;48(8):1105–16. doi: 10.1016/j.neuropharm.2005.03.016. [DOI] [PubMed] [Google Scholar]
  95. McPartland JM, Duncan M, Di Marzo V, Pertwee RG. Are cannabidiol and Δ9-tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. Br J Pharmacol. 2015;172(3):737–53. doi: 10.1111/bph.12944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Mechoulam R, Ben-Shabat S. From gan-zi-gun-nu to anandamide and 2-arachidonoylglycerol: the ongoing story of cannabis. Nat Prod Rep. 1999;16(2):131–43. doi: 10.1039/a703973e. [DOI] [PubMed] [Google Scholar]
  97. Mechoulam R, Ben-Shabat S, Hanuš L, Ligumsky M, Kaminski NE, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol. 1995;50(1):83–90. doi: 10.1016/0006-2952(95)00109-d. [DOI] [PubMed] [Google Scholar]
  98. Mechoulam R, Parker LA. The endocannabinoid system and the brain. Annu Rev Psychol. 2013;64:21–47. doi: 10.1146/annurev-psych-113011-143739. [DOI] [PubMed] [Google Scholar]
  99. Mechoulam R, Shani A, Edery H, Grunfeld Y. Chemical basis of hashish activity. Science. 1970;169(3945):611–12. doi: 10.1126/science.169.3945.611. [DOI] [PubMed] [Google Scholar]
  100. Mechoulam R, Shvo Y. Hashish—I: The structure of cannabidiol. Tetrahedron. 1963;19(12):2073–78. doi: 10.1016/0040-4020(63)85022-x. [DOI] [PubMed] [Google Scholar]
  101. Melis M, Pistis M, Perra S, Muntoni AL, Pillolla G, Gessa GL. Endocannabinoids mediate presynaptic inhibition of glutamatergic transmission in rat ventral tegmental area dopamine neurons through activation of CB1 receptors. J Neurosci. 2004;24(1):53–62. doi: 10.1523/JNEUROSCI.4503-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Mencher SK, Wang LG. Promiscuous drugs compared to selective drugs (promiscuity can be a virtue) BMC Clin Pharmacol. 2005;5:3. doi: 10.1186/1472-6904-5-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Moreira FA, Aguiar DC, Guimarães FS. Anxiolytic-like effect of cannabidiol in the rat Vogel conflict test. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30(8):1466–71. doi: 10.1016/j.pnpbp.2006.06.004. [DOI] [PubMed] [Google Scholar]
  104. Morgan CJA, Das RK, Joye A, Curran HV, Kamboj SK. Cannabidiol reduces cigarette consumption in tobacco smokers: preliminary findings. Addict Behav. 2013;38(9):2433–36. doi: 10.1016/j.addbeh.2013.03.011. [DOI] [PubMed] [Google Scholar]
  105. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1993;365(6441):61–65. doi: 10.1038/365061a0. [DOI] [PubMed] [Google Scholar]
  106. Nestler EJ, Carlezon WA., Jr The mesolimbic dopamine reward circuit in depression. Biol Psychiatry. 2006;59(12):1151–59. doi: 10.1016/j.biopsych.2005.09.018. [DOI] [PubMed] [Google Scholar]
  107. Onaivi ES, Green MR, Martin BR. Pharmacological characterization of cannabinoids in the elevated plus maze. J Pharmacol Exp Ther. 1990;253(3):1002–9. [PubMed] [Google Scholar]
  108. O’Sullivan SE, Sun Y, Bennett AJ, Randall MD, Kendall DA. Time-dependent vascular actions of cannabidiol in the rat aorta. Eur J Pharmacol. 2009;612(1–3):61–68. doi: 10.1016/j.ejphar.2009.03.010. [DOI] [PubMed] [Google Scholar]
  109. Pandolfo P, Silveirinha V, dos Santos-Rodrigues A, Venance L, Ledent C, et al. Cannabinoids inhibit the synaptic uptake of adenosine and dopamine in the rat and mouse striatum. Eur J Pharmacol. 2011;655(1–3):38–45. doi: 10.1016/j.ejphar.2011.01.013. [DOI] [PubMed] [Google Scholar]
  110. Panlilio LV, Justinova Z, Goldberg SR. Inhibition of FAAH and activation of PPAR: new approaches to the treatment of cognitive dysfunction and drug addiction. Pharmacol Ther. 2013;138(1):84–102. doi: 10.1016/j.pharmthera.2013.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Parker LA, Burton P, Sorge RE, Yakiwchuk C, Mechoulam R. Effect of low doses of Δ9-tetrahydrocannabinol and cannabidiol on the extinction of cocaine-induced and amphetamine-induced conditioned place preference learning in rats. Psychopharmacology. 2004;175(3):360–66. doi: 10.1007/s00213-004-1825-7. [DOI] [PubMed] [Google Scholar]
  112. Perras C. Sativex for the management of multiple sclerosis symptoms. Issues Emerg Health Technol. 2005;72:1–4. [PubMed] [Google Scholar]
  113. Pertwee RG. GPR55: a new member of the cannabinoid receptor clan? Br J Pharmacol. 2007;152(7):984–86. doi: 10.1038/sj.bjp.0707464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: Δ9-tetrahydrocannabinol, cannabidiol and Δ9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153(2):199–215. doi: 10.1038/sj.bjp.0707442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Pertwee RG, Ross RA, Craib SJ, Thomas A. (−)-Cannabidiol antagonizes cannabinoid receptor agonists and noradrenaline in the mouse vas deferens. Eur J Pharmacol. 2002;456(1–3):99–106. doi: 10.1016/s0014-2999(02)02624-9. [DOI] [PubMed] [Google Scholar]
  116. Pompeiano M, Palacios JM, Mengod G. Distribution and cellular localization of mRNA coding for 5-HT1A receptor in the rat brain: correlation with receptor binding. J Neurosci. 1992;12(2):440–53. doi: 10.1523/JNEUROSCI.12-02-00440.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Potenza MN, Sofuoglu M, Carroll KM, Rounsaville BJ. Neuroscience of behavioral and pharmacological treatments for addictions. Neuron. 2011;69(4):695–712. doi: 10.1016/j.neuron.2011.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Qin N, Neeper MP, Liu Y, Hutchinson TL, Lubin ML, Flores CM. TRPV2 is activated by cannabidiol and mediates CGRP release in cultured rat dorsal root ganglion neurons. J Neurosci. 2008;28(24):6231–38. doi: 10.1523/JNEUROSCI.0504-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Rahimi A, Hajizadeh Moghaddam A, Roohbakhsh A. Central administration of GPR55 receptor agonist and antagonist modulates anxiety-related behaviors in rats. Fundam Clin Pharmacol. 2015;29(2):185–90. doi: 10.1111/fcp.12099. [DOI] [PubMed] [Google Scholar]
  120. Rakhshan F, Day TA, Blakely RD, Barker EL. Carrier-mediated uptake of the endogenous cannabinoid anandamide in RBL-2H3 cells. J Pharmacol Exp Ther. 2000;292(3):960–67. [PubMed] [Google Scholar]
  121. Ren Y, Whittard J, Higuera-Matas A, Morris CV, Hurd YL. Cannabidiol, a nonpsychotropic component of cannabis, inhibits cue-induced heroin seeking and normalizes discrete mesolimbic neuronal disturbances. J Neurosci. 2009;29(47):14764–69. doi: 10.1523/JNEUROSCI.4291-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Resstel LBM, Joca SRL, Moreira FA, Corrêa FMA, Guimarães FS. Effects of cannabidiol and diazepam on behavioral and cardiovascular responses induced by contextual conditioned fear in rats. Behav Brain Res. 2006;172(2):294–98. doi: 10.1016/j.bbr.2006.05.016. [DOI] [PubMed] [Google Scholar]
  123. Resstel LBM, Tavares RF, Lisboa SFS, Joca SRL, Corrêa FMA, Guimarães FS. 5-HT 1A receptors are involved in the cannabidiol-induced attenuation of behavioural and cardiovascular responses to acute restraint stress in rats. Br J Pharmacol. 2009;156(1):181–88. doi: 10.1111/j.1476-5381.2008.00046.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Riedel G, Fadda P, McKillop-Smith S, Pertwee RG, Platt B, Robinson L. Synthetic and plant-derived cannabinoid receptor antagonists show hypophagic properties in fasted and non-fasted mice. Br J Pharmacol. 2009;156(7):1154–66. doi: 10.1111/j.1476-5381.2008.00107.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Riegel AC, Lupica CR. Independent presynaptic and postsynaptic mechanisms regulate endocannabinoid signaling at multiple synapses in the ventral tegmental area. J Neurosci. 2004;24(49):11070–78. doi: 10.1523/JNEUROSCI.3695-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Rinaldi-Carmona M, Barth F, Congy C, Martinez S, Oustric D, et al. SR147778 [5-(4-Bromophenyl)-1-(2,4-dichlorophenyl)-4-ethyl-N-(1-piperidinyl)-1H-pyrazole-3-carboxamide], a new potent and selective antagonist of the CB1 cannabinoid receptor: biochemical and pharmacological characterization. J Pharmacol Exp Ther. 2004;310(3):905–14. doi: 10.1124/jpet.104.067884. [DOI] [PubMed] [Google Scholar]
  127. Robbe D, Kopf M, Remaury A, Bockaert J, Manzoni OJ. Endogenous cannabinoids mediate long-term synaptic depression in the nucleus accumbens. PNAS. 2002;99(12):8384–88. doi: 10.1073/pnas.122149199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Roberts DC, Corcoran ME, Fibiger HC. On the role of ascending catecholaminergic systems in intravenous self-administration of cocaine. Pharmacol Biochem Behav. 1977;6(6):615–20. doi: 10.1016/0091-3057(77)90084-3. [DOI] [PubMed] [Google Scholar]
  129. Robson P. Abuse potential and psychoactive effects of δ-9-tetrahydrocannabinol and cannabidiol oromucosal spray (Sativex), a new cannabinoid medicine. Expert Opin Drug Saf. 2011;10(5):675–85. doi: 10.1517/14740338.2011.575778. [DOI] [PubMed] [Google Scholar]
  130. Ross RA. The enigmatic pharmacology of GPR55. Trends Pharmacol Sci. 2009;30(3):156–63. doi: 10.1016/j.tips.2008.12.004. [DOI] [PubMed] [Google Scholar]
  131. Russo EB, Burnett A, Hall B, Parker KK. Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem Res. 2005;30(8):1037–43. doi: 10.1007/s11064-005-6978-1. [DOI] [PubMed] [Google Scholar]
  132. Russo EB, Guy GW. A tale of two cannabinoids: the therapeutic rationale for combining tetrahydrocannabinol and cannabidiol. Med Hypotheses. 2006;66(2):234–46. doi: 10.1016/j.mehy.2005.08.026. [DOI] [PubMed] [Google Scholar]
  133. Russo SJ, Nestler EJ. The brain reward circuitry in mood disorders. Nat Rev Neurosci. 2013;14(9):609–25. doi: 10.1038/nrn3381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Ryan D, Drysdale AJ, Pertwee RG, Platt B. Differential effects of cannabis extracts and pure plant cannabinoids on hippocampal neurones and glia. Neurosci Lett. 2006;408(3):236–41. doi: 10.1016/j.neulet.2006.09.008. [DOI] [PubMed] [Google Scholar]
  135. Ryberg E, Larsson N, Sjögren S, Hjorth S, Hermansson N-O, et al. The orphan receptor GPR55 is a novel cannabinoid receptor. Br J Pharmacol. 2007;152(7):1092–101. doi: 10.1038/sj.bjp.0707460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Santos CJPA, Stern CAJ, Bertoglio LJ. Attenuation of anxiety-related behaviour after the antagonism of transient receptor potential vanilloid type 1 channels in the rat ventral hippocampus. Behav Pharmacol. 2008;19(4):357–60. doi: 10.1097/FBP.0b013e3283095234. [DOI] [PubMed] [Google Scholar]
  137. Scherma M, Panlilio LV, Fadda P, Fattore L, Gamaleddin I, et al. 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. 2008;327(2):482–90. doi: 10.1124/jpet.108.142224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Schoedel KA, Chen N, Hilliard A, White L, Stott C, et al. A randomized, double-blind, placebo-controlled, crossover study to evaluate the subjective abuse potential and cognitive effects of nabiximols oromucosal spray in subjects with a history of recreational cannabis use. Hum Psychopharmacol. 2011;26(3):224–36. doi: 10.1002/hup.1196. [DOI] [PubMed] [Google Scholar]
  139. Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, et al. 2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain. Biochem Biophys Res Commun. 1995;215(1):89–97. doi: 10.1006/bbrc.1995.2437. [DOI] [PubMed] [Google Scholar]
  140. Terzian ALB, Aguiar DC, Guimarães FS, Moreira FA. Modulation of anxiety-like behaviour by Transient Receptor Potential Vanilloid Type 1 (TRPV1) channels located in the dorsolateral periaqueductal gray. Eur Neuropsychopharmacol. 2009;19(3):188–95. doi: 10.1016/j.euroneuro.2008.11.004. [DOI] [PubMed] [Google Scholar]
  141. Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG. Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol. 2007;150(5):613–23. doi: 10.1038/sj.bjp.0707133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Touw M. The religious and medicinal uses of cannabis in China, India and Tibet. J Psychoact Drugs. 1981;13(1):23–34. doi: 10.1080/02791072.1981.10471447. [DOI] [PubMed] [Google Scholar]
  143. Tudge L, Williams C, Cowen PJ, McCabe C. Neural effects of cannabinoid CB1 neutral antagonist tetrahydrocannabivarin on food reward and aversion in healthy volunteers. Int J Neuropsychopharmacol. 2015;18(6):pyu094. doi: 10.1093/ijnp/pyu094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Twitchell W, Brown S, Mackie K. Cannabinoids inhibit N- and P/Q-type calcium channels in cultured rat hippocampal neurons. J Neurophysiol. 1997;78(1):43–50. doi: 10.1152/jn.1997.78.1.43. [DOI] [PubMed] [Google Scholar]
  145. Volkow ND, Morales M. The brain on drugs: from reward to addiction. Cell. 2015;162(4):712–25. doi: 10.1016/j.cell.2015.07.046. [DOI] [PubMed] [Google Scholar]
  146. Wade DT, Makela P, Robson P, House H, Bateman C. Do cannabis-based medicinal extracts have general or specific effects on symptoms in multiple sclerosis? A double-blind, randomized, placebo-controlled study on 160 patients. Mult Scler. 2004;10(4):434–41. doi: 10.1191/1352458504ms1082oa. [DOI] [PubMed] [Google Scholar]
  147. Ward SJ, Raffa RB. Rimonabant redux and strategies to improve the future outlook of CB1 receptor neutral-antagonist/inverse-agonist therapies. Obesity. 2011;19(7):1325–34. doi: 10.1038/oby.2011.69. [DOI] [PubMed] [Google Scholar]
  148. Wilson RI, Nicoll RA. Endogenous cannabinoids mediate retrograde signalling at hippocampal synapses. Nature. 2001;410(6828):588–92. doi: 10.1038/35069076. [DOI] [PubMed] [Google Scholar]
  149. Wise RA, Bozarth MA. Brain mechanisms of drug reward and euphoria. Psychiatr Med. 1985;3(4):445–60. [PubMed] [Google Scholar]
  150. Wolf SA, Bick-Sander A, Fabel K, Leal-Galicia P, Tauber S, et al. Cannabinoid receptor CB1 mediates baseline and activity-induced survival of new neurons in adult hippocampal neurogenesis. Cell Commun Signal CCS. 2010;8:12. doi: 10.1186/1478-811X-8-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Xi Z-X, Peng X-Q, Li X, Song R, Zhang H, et al. Brain cannabinoid CB2 receptors modulate cocaine’s actions in mice. Nat Neurosci. 2011;14(9):1160–66. doi: 10.1038/nn.2874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Xiong W, Cui T, Cheng K, Yang F, Chen S-R, et al. Cannabinoids suppress inflammatory and neuropathic pain by targeting α3 glycine receptors. J Exp Med. 2012;209(6):1121–34. doi: 10.1084/jem.20120242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Yoshimoto K, McBride WJ. Regulation of nucleus accumbens dopamine release by the dorsal raphe nucleus in the rat. Neurochem Res. 1992;17(5):401–7. doi: 10.1007/BF00969884. [DOI] [PubMed] [Google Scholar]
  154. Zanelati TV, Biojone C, Moreira FA, Guimarães FS, Joca SRL. Antidepressant-like effects of cannabidiol in mice: possible involvement of 5-HT1A receptors. Br J Pharmacol. 2010;159(1):122–28. doi: 10.1111/j.1476-5381.2009.00521.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Zhang H-Y, Gao M, Liu Q-R, Bi G-H, Li X, et al. Cannabinoid CB2 receptors modulate midbrain dopamine neuronal activity and dopamine-related behavior in mice. PNAS. 2014;111(46):E5007–15. doi: 10.1073/pnas.1413210111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Zuardi AW. History of cannabis as a medicine: a review. Rev Bras Psiquiatr. 2006;28(2):153–57. doi: 10.1590/s1516-44462006000200015. [DOI] [PubMed] [Google Scholar]
  157. Zuardi AW, Cosme RA, Graeff FG, Guimarães FS. Effects of ipsapirone and cannabidiol on human experimental anxiety. J Psychopharmacol. 1993;7(1 Suppl):82–88. doi: 10.1177/026988119300700112. [DOI] [PubMed] [Google Scholar]
  158. Zuardi AW, Shirakawa I, Finkelfarb E, Karniol IG. Action of cannabidiol on the anxiety and other effects produced by Δ9-THC in normal subjects. Psychopharmacology. 1982;76(3):245–50. doi: 10.1007/BF00432554. [DOI] [PubMed] [Google Scholar]

RESOURCES