Next Article in Journal
Perampanel in Brain Tumor-Related Epilepsy: A Systematic Review
Next Article in Special Issue
Risk of Motor Vehicle Collisions and Culpability among Older Drivers Using Cannabis: A Meta-Analysis
Previous Article in Journal
Responsive Neurostimulation of the Anterior Thalamic Nuclei in Refractory Genetic Generalized Epilepsy: A Case Series
Previous Article in Special Issue
Dual Cannabinoid and Orexin Regulation of Anhedonic Behaviour Caused by Prolonged Restraint Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endocannabinoid System and Exogenous Cannabinoids in Depression and Anxiety: A Review

1
Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
2
McLean Hospital, Belmont, MA 02478, USA
3
Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
4
Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
*
Authors to whom correspondence should be addressed.
Brain Sci. 2023, 13(2), 325; https://doi.org/10.3390/brainsci13020325
Submission received: 6 January 2023 / Revised: 10 February 2023 / Accepted: 11 February 2023 / Published: 14 February 2023
(This article belongs to the Special Issue Cannabis and the Brain: Novel Perspectives and Understandings)

Abstract

:
Background: There is a growing liberalization of cannabis-based preparations for medical and recreational use. In multiple instances, anxiety and depression are cited as either a primary or a secondary reason for the use of cannabinoids. Aim: The purpose of this review is to explore the association between depression or anxiety and the dysregulation of the endogenous endocannabinoid system (ECS), as well as the use of phytocannabinoids and synthetic cannabinoids in the remediation of depression/anxiety symptoms. After a brief description of the constituents of cannabis, cannabinoid receptors and the endocannabinoid system, the most important evidence is presented for the involvement of cannabinoids in depression and anxiety both in human and from animal models of depression and anxiety. Finally, evidence is presented for the clinical use of cannabinoids to treat depression and anxiety. Conclusions: Although the common belief that cannabinoids, including cannabis, its main studied components—tetrahydrocannabinol (THC) and cannabidiol (CBD)—or other synthetic derivatives have been suggested to have a therapeutic role for certain mental health conditions, all recent systematic reviews that we report have concluded that the evidence that cannabinoids improve depressive and anxiety disorders is weak, of very-low-quality, and offers no guidance on the use of cannabinoids for mental health conditions within a regulatory framework. There is an urgent need for high-quality studies examining the effects of cannabinoids on mental disorders in general and depression/anxiety in particular, as well as the consequences of long-term use of these preparations due to possible risks such as addiction and even reversal of improvement.

1. Introduction

There is evidence that humans have used Cannabis sativa since ancient times for recreational and/or medicinal purposes [1]. Although it is still considered illicit by many, multiple countries around the world are softening their position by legalizing the use of these plants or their extracts for medicinal and recreational use. Historically, the identification and isolation of the main psychoactive ingredient of cannabis, Δ9-tetrahydrocannabinol (Δ9-THC) [2] led to the discovery of cannabinoid receptors and, subsequently, led to the identification of endogenous cannabinoids (eCBs), their synthesis, regulation, and the revelation that a complete endogenous endocannabinoid system (ECS) exists, as will be described. Medicinal usage was incorporated for a variety of ailments, ranging from appetite stimulation, insomnia, pain, vomiting, nausea and for other conditions such as cancer, post-traumatic stress disorder, anxiety and depression, to cite a few.
The purpose of this mini-review is to explore the association between depression or anxiety and the dysregulation of the ECS, and the use of phytocannabinoids and synthetic cannabinoids in the remediation of depression/anxiety symptoms. Depression is a widespread debilitating mental illness affecting approximately 300–350 million people globally [3]. Depression is often not an isolated disorder and may be co-morbid with other ailments such as anxiety, cancer, diabetes, stress, pain, and other neuropsychiatric disorders [4,5] for reviews). It is well known that other systems, such as dopamine, serotonin, opioid, adrenergic systems, are also involved in depression and anxiety, but the main focus of the present review is on the endogenous and exogenous cannabinoids in relation to depression and anxiety.
We describe the most important constituents of Cannabis sativa, review the endocannabinoid system, including the different cannabinoid receptors, and the machinery involved in the synthesis, transport and degradation of the endocannabinoids. This is followed by the most established evidence regarding depression/anxiety and ECS, before reviewing the relevant effects of different exogenous cannabinoid preparations in relation to depression and related ailments.

2. Constituents of Cannabis sativa

The chemical nature of the constituents of Cannabis sativa is diverse, with the number of natural compounds totaling 565 [1,6]. Among these, 120 cannabinoids have been isolated and classified into 11 general types, of which, the best known and the most studied are Δ9-trans-tetrahydrocannabinol (THC), and cannabidiol (CBD). THC, first reported by [2], is considered the principal psychoactive component, whereas CBD, first reported by [7], is recognized as a major non-psychotropic constituent of Cannabis sativa [8]. The discovery of these components was followed by the identification of their presumed targets through the cloning of specific cannabinoid receptors. Consequently, the endogenous ligands to these receptors were found in the brain, and called endocannabinoids (eCB), which triggered enormous progress in the exploration of their physiological importance, as well as the relevance of cannabis and its products in many health conditions including mental health [9,10,11].

3. Cannabinoid Receptors

Two receptors have been identified as the main cannabinoid receptors, the CB1 and CB2 receptors. The receptors have been cloned [12,13], established as the primary targets for endogenous and exogenous cannabinoids [10,14,15], and shown to belong to the superfamily of G-protein coupled receptors (GPCRs), characterized by seven transmembrane domains and coupled to G-proteins, notably to Gi/Go proteins [14]. In humans, the genes encoding the CB1 (cnr1) and CB2 (cnr2) receptors are located on the 6q14-q15 and 1p36.11 chromosomes, respectively [10]. The two receptors share ~44% sequence identity and diverge significantly in their distribution. CB1 receptor (CB1R) is highly expressed in different brain regions [16,17,18] and is considered the most abundant GPCR in the brain (reviewed in [10,14,15]). The CB2 receptor (CB2R) is principally expressed in immune tissues, inflammatory cells and in much lower densities in brain [10].

4. Additional Receptors for Cannabinoids

Beside their actions through the CB1 and CB2 receptors, the endocannabinoids, as well as the exogenous cannabinoids, bind also to other receptors [15]. Among these are the transient receptor potential vanilloid 1 (TRPV1) cation channel, peroxisome proliferator-activated receptors (PPARs, [19]), and at least three orphan GPCRs: GPR55, that we have cloned [20] and which has been reported as a cannabinoid receptor [21]; GPR18; and GPR119 [22,23,24]. It is also important to note that many receptor heteromeric complexes have been established in vitro and in vivo, as reviewed in [24,25], such as the CB1R–CB2R heteromers [26], and between CB1R or CB2R and a large variety of other receptors. Notable are the CB1R heteromers with dopamine D2 receptor [27,28], serotonin receptors 5HT1A [29] and 5HT2A [30], adenosine receptor A2A [31], and GPR55 [32]. All of this diversification of signaling through the cannabinoid receptors and the range of heteromers may explain the varied involvement of the cannabinoid system in many pathophysiologic conditions and may be responsible for the variance in response to endogenous and exogenous agonists and pharmacological modulators of the cannabinoid system.

5. The Endocannabinoid System

The endocannabinoid system (ECS) consists of the cannabinoid receptors, the endogenous cannabinoids (eCBs), the enzymes involved in the synthesis and degradation of eCBs and the protein(s) involved in the reuptake of secreted eCBs.

5.1. Endocannabinoids

The primary eCBs are anandamide (AEA) and 2-arachidonoylglycerol (2-AG) (reviewed in [5]). Anandamide (N-arachidonoylethanolamine) was the first endogenous cannabinoid-like substance to be identified [33], which was then followed by 2-AG [34,35]. Both AEA and 2-AG were characterized as endogenous ligands by their capacity to bind and modulate CB1R and CB2R. Other endogenous molecules capable of modulating CB1/CB2 receptors have also been identified [36] and these include 2-AG ether [37], N-arachidonoyl-dopamine [38], O-arachidonoyl ethanolamine, [39], and oleamide [40]. However, the physiological roles of these latter substances have not been completely established yet [5].

5.2. Enzymes Responsible for the Synthesis and Degradation of eCBs

Endocannabinoids are lipophilic molecules synthesized ‘on demand’, meaning synthesis occurs after neuronal activation, from membrane phospholipids, and are released immediately, without storage in vesicles [5]. Both anandamide and 2-AG are produced by post-synaptic neurons (Figure 1A). Anandamide is produced in a two-step process and its levels are regulated by its degradation by the fatty acid amide hydrolase (FAAH) [41]. Also synthesized is 2-AG, in a two-step process involving the action of phospholipase C (PLC), followed by the hydrolytic activity of diacyl glycerol lipase (DAGL) [35]. The levels of 2-AG are essentially regulated by monoacylglycerol lipase (MAGL) [42]. There is a difference however in the location in which both eCBs are degraded. The hydrolysis of 2-AG occurs in presynaptic neurons by MAGL, after CB1R activation, whereas anandamide is hydrolyzed in postsynaptic neurons by FAAH, which may lead to a termination of anandamide action where it is synthesized [5]. In addition to hydrolysis, 2-AG can also be transformed into other bioactive compounds by cyclooxygenase-2 [43] and lipoxygenase [44]. Further, the metabolism of anandamide by lipoxygenase and cyclo-oxygenase enzymes generates oxygenated products that signal through non-cannabinoid targets [5,45].

6. Signaling of the ECS

The mode of action of the eCBs on their receptors (Figure 1) reveals that eCBs synthesized in postsynaptic neurons act pre-synaptically at CB1Rs in a retrograde mode [46]. There are data showing that eCBs can also activate localized CB1Rs post-synaptically [47,48]. In general, activation of CB1 receptors by eCBs decreases the probability of release of neurotransmitters through multiple mechanisms, comprising an inhibition of calcium influx and an activation of potassium channels [46,47,48,49]. These effects are terminated by eCB reuptake followed by degradation; 2AG by MAGL pre-synaptically and AEA by FAAH post-synaptically, as mentioned above. Furthermore, a large proportion (45–48%) of synapses in different brain regions are tripartite [47] (Figure 1B), formed by a pre-synaptic neuron, a post-synaptic neuron, and a glial cell, usually an astrocyte, which expresses CB1R [47]. In response to neuronal activation, eCBs are released by the postsynaptic neurons and activate CB1R expressed on astrocytes. The astrocytes respond with an increase in calcium from intracellular stores leading to the release of gliotransmitters (Figure 1B), capable of modulating the presynaptic and postsynaptic circuit elements [47,50,51]. Through these mechanisms eCBs can inhibit pre-synaptic neurotransmitter release at both GABA and glutamate terminals, thus modulating several neurotransmitter systems [5,47,52]. As illustrated for dopamine neurons, CB1 receptors expressed at presynaptic glutamatergic and GABAergic inputs to DA neurons may facilitate or suppress dopaminergic neuronal activity by modifying such inputs [53,54,55]. CB1 receptors on GABAergic terminals (Figure 2A) can facilitate dopaminergic activity through suppression of the inhibitory input onto GABA receptors present on DA neurons [56,57,58,59], leading to an increase of DA release in areas such as the nucleus accumbens (NAc) of the ventral striatum.
In contrast, CB1 receptors at glutamatergic synapses, suppress excitatory inputs onto the glutamatergic receptors on the DA neurons [57,60,61]. This mechanism involves glutamate release at synapses in prelimbic frontal cortex and in NAc at D1-enriched medium spiny neurons (D1-MSNs) (reviewed in [62]). Activation of the prefrontal glutamate afferents can lead to long-term depression (LTD) of NAc glutamatergic synapses, an effect also involving 2-AG release, presynaptic CB1-receptor and postsynaptic metabotropic glutamate receptor 5 (mGluR5) activation [62,63,64]. Mechanistically (Figure 2B), the activation of mGluR5 by released glutamate leads to the synthesis and release of 2-AG, which then retrogradely activates CB1 receptors to inhibit additional glutamate release (reviewed in [62]). This activation of CB1R by eCBs is usually accompanied by LTD of neurons [65].

7. Reuptake of eCBs

It has been suggested that eCBs are removed from the synaptic space by an uptake mechanism followed by hydrolysis [66], in a process similar to that which happens with other neurotransmitters or neuromodulators such as the well-known example of dopamine re-uptake by dopamine transporter (DAT). However, specific ‘endocannabinoid membrane transporters’, have not yet been identified [5].

8. Cannabinoids in Depression and Anxiety

Because depression and anxiety commonly accompany multiple other disorders, their etiology can be diverse and includes perturbations of multiple systems, in different brain regions; thus, it is difficult to identify the specific roles of cannabinoids in the induction or relief of depression and anxiety. In order to clarify this and simplify the analysis, we propose to approach this review of the role of cannabinoids in depression/anxiety in different sections as follows. First, preclinical data related to cannabinoids and depression- and anxiety-like effects, and stress in animal studies, followed by description of the perturbations of the ECS in humans with depression/anxiety. A third part will highlight the most relevant clinical observations obtained from long-time users of cannabis. Finally, a brief review of the clinical observations resulting from the use of cannabinoids to combat major depression disease (MDD), bipolar disease (BD), and depression/anxiety comorbid with other diseases.

8.1. Cannabinoids and Depression-like Effects in Animal Studies

In preclinical studies using animal model systems, mostly in rodents, cannabinoids, whether exogenous or endogenous, seem to generally have an antidepressant-like effect. However, opposite effects were occasionally observed, due essentially to differences in the dosage, the species, the strains and the experimental design considered. For more detailed reviews see [10,67].
There are a few animal tests used to investigate depression-like and anxiety-like behaviors, such as the forced swim test (FST) and the tail suspension test (TST), to measure manifestations of helplessness, despair and stress engendered by the two tests. The elevated plus maze test (EPM), evaluates the anxiety-like state of the animal subject. The light/dark box is another test that measures the anxiogenic/anxiolytic state of the subject. Another aspect tested is the hedonia/anhedonia level after treatments, which can be assessed using the sucrose preference test (SPT). All these tests can monitor the status at basal level, after administering a drug or after an experimental maneuver, such as after chronic unpredictable mild stress (CUS).
Several lines of evidence suggest that acute enhancement of CB1R activity results in antidepressant- and anxiolytic-like effects in rodents (Table 1). For example, activation of CB1R by administration of its agonists HU210 or oleamide resulted in antidepressant-like action in the rat FST model [68]. Similar antidepressant and/or anxiolytic effects were observed after acute activation of CB1R by THC [69,70] or other cannabinoids, such as the synthetic agonists WIN55, 212–2 [71], CP55, 940 [72], as well as the endogenous AEA [73,74]. Other cannabinoid agonists or partial agonists, endogenous and exogenous, also showed similar effects (reviewed in [10,67]). The efficacy of these cannabinoid compounds was as good as some known antidepressants with which they were compared. In most cases, these positive effects on anxiety and depression were observed at relatively low doses of cannabinoids activating CB1R, whereas higher doses were often pro-depressive and anxiogenic [69,75,76,77,78,79]. This observation could mirror the biphasic effects usually seen in humans [10,69]. There are, however, some reported differences, which were essentially explained by species (mouse vs. rat) and strain differences, the brain region injected or studied, the test used, or other experimental differences [10,67].

8.2. Evidence for the Involvement of the ECS in Depression, Anxiety, and Stress

The involvement of the CB1R in the above-described effects of cannabinoids was usually verified by blockade of the receptor, using an antagonist/inverse agonist, such as the most known rimonabant [86] or AM251 [86]. However, opposing results have also been shown by some studies in which it was reported that the antagonist SR141716A (structurally comparable to rimonabant) and AM251 exerted antidepressant-like effects in animal models [87,88,89,90]. In the case of rimonabant, the effects were similar to those of antidepressant fluoxetine in various animal models of depression [90]. Moreover, such an effect was absent from the gene-deleted CB1−/− mouse treated with AM251, indicating a role for the CB1 receptor in depression. The mechanism underlying the antidepressant-like effects of rimonabant and other antagonists or inverse agonists remains to be determined, especially since rimonabant in humans has shown pro-depressive, pro-anxiogenic and even ideations of suicide, as will be described later. In contrast, multiple animal studies have shown that the blockade of CB1R signaling in rodents results in anhedonia-like behavior [91], and heightened levels of basal anxiety [92,93,94], and in general, an increased tendency to develop a passive coping response to stress [95,96]. In these latter studies, an increase in the hypothalamic–pituitary-adrenal axis (HPA axis) activity under both basal conditions and following exposure to stress was also observed [86,95,96,97].
Models of depression and anxiety involving stress have shown that the function of the ECS is reduced. For example, CB1R has been shown to be decreased in several brain structures implicated in depression in rats exposed to chronic unpredictable stress (CUS) [98,99,100]. When tested, the levels of eCBs, notably AEA, were reduced in different brain structures in studies of the CUS model of depression [97,98,99,100,101,102], demonstrating that chronic stress paradigms result invariably in a loss of AEA signaling. In addition, chronic stress exposure dramatically decreased the aptitude of CB1R to inhibit neurotransmitter release in regions involved in depression, such as the striatum and hypothalamus [103,104,105]. Additional evidence for the role of the ECS, and precisely the CB1R, in depression in preclinical studies came from knock-out mouse models in which CB1R (CB1R-KO) was eliminated. To simplify, studies from this model showed that decrease or elimination of CB1R or its signaling resembles the effects of antagonists, in that it is pro-depressive and pro-anxiogenic overall.
Other evidence derives from the use of a fatty acid amide hydrolase (FAAH) inhibitor or inhibitor of reuptake of eCBs. Chronic administration of the FAAH inhibitor URB597, which would increase AEA levels, has been shown to reduce anxiety-like effects and to display antidepressant-like actions in a CUS rat model; effects that were prevented by a CB1R-selective antagonist SR141761A [106]. This effect was not always observed [107], as URB597, an irreversible FAAH inhibitor, failed to demonstrate antidepressant-like actions in either the FST or TST. The use of other FAAH inhibitors or the use of FAAH gene deletion (FAAH-KO) model mouse, however, showed clearly that inhibition (or ablation) of FAAH was anxiolytic and anti-depressive (reviewed in [68]). These results are corroborated by the use of an inhibitor for eCB uptake, AM404. Accumulation of eCBs following administration of the uptake inhibitor AM404 elicited an antidepressant-like effect, presumably by indirect stimulation of the CB1 receptors by eCBs [68].
Thus, most preclinical studies underline an important role for the ECS in anxiety- and depression-like behaviors, and that the ECS seems to be compromised in the animal models of depression, anxiety, and stress exposure.

8.3. Perturbations of the ECS in Humans with Depression

In humans, there are multiple lines of evidence suggesting a role for the ECS in depression, anxiety, and stress. Most of the available evidence for perturbations affecting ECS in depression and anxiety derive from two major sources: post-mortem tissues and clinical measurements of eCB levels, notably AEA and 2AG, in the blood or cerebrospinal fluid (CSF) of patients. The other observations are related to cannabis use for recreational or self-medication purposes, which will be treated independently.
Information pertaining to the role of ECS in human depression, anxiety and stress is relatively scarce, and not completely validated, as reviewed in [67,108,109]. From post-mortem studies, an upregulation of CB1R and its G-protein signaling has been observed in the dorsolateral prefrontal cortex of depressed suicide victims [110]. Elevated levels of the eCBs AEA and 2-AG, and CB1R-mediated G-protein signaling in the prefrontal cortex of alcoholic suicide victims were also observed [111]. The up-regulation of CB1R coupling to G-proteins, specifically to Gαo, and not Gαi or Gαz protein subunits, has also been observed in the prefrontal cortex of depressed suicide victims [112]. It has been revealed, however, that this up-regulated CB1R/Gαo functionality might be due in part to the administration of antidepressants [67,112]. An interesting point is that the fluctuations of the eCBs seem to be linked to the gravity of depression [67]. Increases in serum levels of 2-AG and/or AEA has been observed in patients with minor depression, whereas patients with major depression showed reduced serum levels of 2-AG and/or AEA. Similarly, CB1R levels seem to depend on the severity of the depression. For example, CB1R-immunopositive glial cells in the grey matter were decreased in major depression, whereas no evidence of an altered density of CB1R immunopositive cells in schizophrenia or bipolar disorder was found [113]. The evaluation of the circulating levels of eCBs in body fluids may give an indication as to the role of the ECS in these disorders, although, as mentioned above, the severity of the disease and pre-treatment with other drugs, including antidepressants, may play a role in the final levels quantified. For example, it has been reported that plasma 2-AG levels, as well as CB1 and CB2 mRNA levels, are elevated in the lymphocytes of osteoarthritic patients, with a positive correlation between 2-AG levels, pain and depression [5]. In a recent systematic review of different studies tackling this matter [108], it was reported that differential effects of stress, depression, and anxiety symptoms may modulate endogenous levels of the eCBs. For example, acute psychological stress increased AEA and other fatty acid ethanolamides in healthy volunteers [114], whereas, in a study involving PTSD patients [115] there was a hypoactive eCB system, notably with decreased AEA and 2-AG, among others, in the plasma of the participants after physical and psychosocial stress. In another study involving patients with treatment-resistant depression or bipolar disorder, before and after electroconvulsive treatment [116], it was noted that the treatment increased AEA in the CSF of patients. In a cohort of students who were carriers of the A allele of the FAAH enzyme, characterized by increased basal AEA, this elevation was shown to facilitate fear extinction and ameliorate extinction of recall [117]. Moreover, it was suggested that the polymorphism in the FAAH gene was associated with bipolar disorder and major depression [118] which is in line with the importance of FAAH activity in depression and suicidal behavior [111].
Reducing endocannabinoid signaling in humans is usually sufficient to produce depressive symptoms [119,120]. As an example, the CB1 receptor antagonist/inverse agonist, rimonabant, has been used as a treatment for obesity in humans. However, its administration led to severe symptoms of anxiety and depression in a significant proportion of individuals [121,122], which resulted in the removal of rimonabant from the market [119]. Another line of evidence comes from genetic studies that showed that some genetic polymorphisms in CB1R and CB2R may be associated with major depression and bipolar disorder, as reviewed in [119]. Some of these genetic studies also showed that single nucleotide polymorphisms (SNPs) in the CB1R can increase the vulnerability to the development of a depressive episode following exposure to life stress [123], and confer an increased risk of antidepressant resistance [124]. It has also been found that SNPs in the CB1 receptor gene can result in blunted neuronal activation in response to rewarding stimuli [124]. A significant increase in the frequency of these CB1 receptor SNPs in patients with mood disorders was also observed [118].
From these studies, it is clear that perturbations of the ECS play a role in depression, anxiety and response to stress. It also indicates that, in general, increasing eCB levels would be beneficial in some neuropsychiatric disorders, including MDD and probably chronic anxiety. This suggests also that blocking the degradation of the eCBs and/or activating the ECS may ameliorate these particular conditions.

9. Clinical Observations Obtained from Long-Time Cannabis Users

Recreational and potential medicinal usage of cannabis and derivatives is based on their perceived acute properties of inducing euphoria, relaxation and analgesia, as well as promoting sleep, appetite stimulation and other effects such as relief of chronic pain, nausea and vomiting. However, multiple adverse effects have been described in long-term users of cannabis, including possible development of psychosis [125] and increased depression and anxiety [108,126,127,128]. Evidence suggests that both short- and long-term use of cannabis heighten the risks for psychiatric illness. A clear example is the usual cognitive impairment observed in both acute and chronic cannabis users, which includes impaired short-term memory, motor coordination and control, sleep, executive functioning, as well as altered judgement, which, in sum, affect most aspects of regular day-to-day life of the users [125,129,130,131,132]. Additionally, high doses of cannabis, and more importantly, high content THC in some engineered cannabis plants and preparations can induce paranoia and psychosis even after single doses or short-term cannabis use and may even induce these in the long-term, structural brain modifications in younger brains [133,134]. Furthermore, chronic cannabis use has been associated with a higher risk of developing cannabis use disorder (CUD) and may lead to other substance use disorders (SUDs) [130,135,136,137], including heightened functional impairment of normal daily activities comparable to the harms observed with any SUD, as reviewed in [135,138,139]. A large proportion of long-term users would transition to CUD and most experience strong withdrawal symptoms after cessation [128,140,141,142,143], including irritability, anger or aggression, nervousness or anxiety, sleep disruption, decreased appetite, restlessness, or depressed mood, all of which could promote further drug seeking and relapse. Clinically, 50–95% of heavy users may suffer from some symptoms of cannabis withdrawal [140,141,142], with a meta-analysis reporting an overall prevalence of 47% [143]. Moreover, it has also been shown that long-term use of cannabis may lead to the use of other illicit drugs, which is reinforced by increased frequency and earlier onset of use [144], thus increasing the risk of SUDs and closely aligned mental illnesses, notably depression and anxiety. In a paradoxical twist, the use of cannabis to self-medicate for depression and anxiety may in fact lead to CUD (or SUD), with individuals suffering with a mental illness generating higher dependence on cannabis as a coping strategy for the management of these symptoms and consequently augmenting the risk of developing CUD (or SUD), which in turn may lead to an augmented risk of exacerbating severe depression and anxiety. These effects can be worsened by different factors, such as an earlier age of onset of use, the quantity and potency, the frequency, as well as the period of use and percentage of THC in the preparation [11,145]. In any case, these effects are essentially due to the actions of THC acting as a partial agonist at CB1R [146,147]. In contrast with THC, the use of CBD, the second most abundant cannabinoid in cannabis, has been shown to be devoid of any direct psychoactive behavioral effect in animal studies and, notably, had no rewarding effect, thus negating its potential for abuse as an addictive substance [148,149]. It has been shown that CBD can counter many of the actions of THC and may even provide a certain degree of protection against some, but not all, psychoactive and other deleterious properties of THC [11,145].
In relation to depression and anxiety, a higher incidence of depression and anxiety is usually the norm among long-term users of cannabis, although this could be due to other confounding and comorbid factors. For example, as reviewed in [128], in major depressive disorder (MDD) and bipolar disorder (BD), evidence generally reveals a harmful outcome of long-term cannabis use. Cannabis use was correlated with a higher risk of MDD diagnosis, and earlier onset of cannabis use was correlated with shorter time to MDD diagnosis [128,150]. This positive correlation between long-term cannabis use and depression or anxiety seems to be a consistent finding, although some other studies indicate this correlation disappears when controlling for multiple confounding factors, such as the co-use of other drugs, sex, age, genetic traits and education level, among others [128,151,152,153]. Similarly, evidence indicates the harmful effects of long-term cannabis use in bipolar disorder (BD), with frequent cannabis use being associated with heightened risk of BD onset [154] and/or exacerbation of BD symptomology [154,155,156]. It is worth noting that these effects of long-time use of cannabis are in opposition to the effects of acute cannabis use, which seems to lead to an improvement in BD symptoms [157]. As for MDD, there are studies that found no statistically significant correlation between cannabis use and BD consequences when other factors were considered [157,158]. Anxiety symptoms seem to be higher in cannabis users compared with non-users [159] and are enhanced with the earlier onset of cannabis use [159,160,161,162]. Another condition, which can be related to anxiety and depression, is post-traumatic stress disorder (PTSD), for which cannabis is being used either as self-medication or doctor-prescribed medication. While those undergoing acute or short time treatment, mostly medically assisted, seem to show some benefits in lowering anxiety levels and ameliorating the quality of life, it has been shown that daily cannabis use is positively associated with increased severity of PTSD symptoms [163]. Most of the evidence supports an association of long-time cannabis use with a higher risk of PTSD symptoms and heightened negative affect [128,163,164].
In sum, the acute use of cannabis may offer relief from various symptoms linked to anxiety, stress and depression. However, the long-term consequences of cannabis use seem to have higher potential for harmful outcomes for these conditions. Nevertheless, other confounding factors may play a role in heightening these risks. Indeed, as recently reported [109,164], the World Health Organization (WHO, [3]) and the National Academies of Science, Engineering, and Medicine [165] concluded that the effects of cannabis use on depression seem to be limited and slightly increase the risk of developing depression. Nevertheless, a systematic review [166] that appraised 44 previous systematic reviews, including 1053 different studies covering a broad spectrum of negative health outcomes directly connected to cannabis use, concluded that a clear association exists between cannabis use and psychosis, affective disorders, anxiety, sleep disorders, cognitive failures, respiratory adverse events, cancer, cardiovascular outcomes, and gastrointestinal disorders. Several studies have concluded that cannabis use during adolescence had a strong impact on risk for psychosis and incidence of schizophrenia as well as a causal association with poor school performance, lower educational attainment and early school dropouts, as reviewed in [166]. Many studies have also reported that cannabis use increased the risk for developing MDD and BD, as well as for maintaining high levels of anxiety over time, as reviewed in [128,166]. All these diverse harms that ensue from cannabis use have led different studies to conclude that cannabis use and misuse constitute a highly relevant public health challenge [166].

10. Clinical Use of Cannabinoids to Treat Depression, Anxiety, and Stress

Although the long-term effects of cannabis and its derivatives are not completely known, except on the part of the long-time users of cannabis themselves, the medical potential for cannabis and cannabis-based products have been suggested by multiple observational, open-label studies, and a few RCTs [108,128] (Table 2). The presumed medicinal indications cover a large array of conditions, including chronic pain, epilepsy, insomnia, cancer-associated pain, vomiting and nausea in cancer and HIV patients, PTSD, Parkinson’s disease (PD), depression, anxiety, and stress, among others [164]. These studies, however, contain multiple weaknesses, as several recent systematic reviews and meta-analyses have found [108,109,128]. Nevertheless, among the multiple constituents of cannabis, THC and CBD have been the most subject to different investigations in animal models and trials in humans. There are also some studies of synthetic cannabinoids or endogenous cannabinoids [5,108,128,167,168,169]. In general, cannabis and its derivatives seem to be well tolerated by humans and animals alike when used at relatively low doses and during short time periods [168,169,170]. Under these conditions, they are usually linked to many positive therapeutic effects for a wide range of symptoms and diseases [164,170]. Briefly described are the therapeutic effects reported for THC, CBD, and some synthetic cannabinoids and, for each, the available data related to effects on anxiety, stress and depression are presented.

10.1. Δ9-Tetrahydrocannabinol

The biphasic effect reported in animal studies, with low doses reducing anxiety and high doses producing anxiogenic-like and antidepressant-like effects [69,70,71,72] are also seen in humans. THC has demonstrated biphasic, dose-dependent effects on anxiety in healthy adults [173,174,175,176,177,178]. Neuroimaging data have also confirmed that THC can both increase [175] and decrease [174] emotional arousal/processing of negative stimuli at specific doses [178]. Certain THC doses globally induced anxiogenic effects in healthy individuals, although in this study half of the subjects were recreational users of cannabis [108,179]. However, due to the credible harmful effects over the long-term, such as addictive potential and the induction of anxiety and depression, multiple meta-analyses and reviews warn against the use of high doses of pure THC or cannabis-based preparations containing THC for long periods of time [108,128].
Other therapeutic effects of THC reveal stimulation of appetite, and it is used for this effect to treat patients with anorexia and cachexia of advanced cancer [180]. THC is also used in chemotherapy patients due to its effect of reducing nausea and vomiting [181,182]. THC has been shown to reduce acute and chronic pain (for a review see [5,170,183], including neuropathic pain [184,185,186]. THC has also been shown to help overcome some negative symptoms linked to PTSD in war veterans [167,187,188]. There are two synthetic analogs of THC approved by the US Food and Drug Administration (FDA) that could be prescribed for chemotherapy-induced nausea and vomiting: nabilone and dronabinol (reviewed in [189]). Nabilone has been shown to be effective in reducing vomiting and nausea [190], whereas dronabinol is currently being assessed for its analgesic properties in patients with breast cancer and in patients with chronic pain [5,167,189], anorexia of HIV/AIDS and cancer patients [191,192]. The antiemetic effects, consisting of the easing of nausea and/or vomiting have also been observed in animal models [189,193,194,195]. Amelioration of activity and motor impairment have been observed in animal models of Parkinson’s Disease (PD) [196] and Amyotrophic Lateral Sclerosis (ALS) [197].

10.2. Cannabidiol

Multiple positive effects have been linked to CBD (for recent reviews, see [167,189,198]. These include neuroprotective properties and reported effects as an anxiolytic and antipsychotic agent. Other potential therapeutic benefits attached to CBD include amelioration of symptoms in disorders such as PTSD, bipolar disorder, epilepsy, social phobia, schizophrenia, Parkinson’s disease and more [189]. Preclinical animal models suggest that CBD has beneficial effects by itself as well as reducing or abolishing some negative THC-induced effects, notably at long-term [108,128]. It also shows some benefit in reducing nausea and vomiting [199] and has an anticonvulsant effect, which is a promising potential for use, especially for children with epileptic syndromes [189,200].
Unlike THC, CBD has no psychoactive effect, but can in fact counter the psychoactive effects of THC and other negative effects, such as inhibiting the THC-induced cognitive impairment in preclinical studies involving non-human primates [201]. In depression, anxiety, and stress, CBD has constantly been shown to induce anxiolytic and antidepressive potential [108,128,178]. CBD has also been shown to counter many of the effects of THC, including THC-induced anxiogenic effects [173,174,175,176,178,202]. Healthy participants have also reported decreased levels of subjective anxiety 90-min post administration of CBD. Some anxiolytic effects of CBD have been seen to be similar to the post-stress anxiolytic effects of isapirone, a selective 5-HT1A receptor partial agonist known for its antidepressant and anxiolytic effects [178,202]. The positive effects of CBD have also been shown in multiple other studies, such as those with healthy volunteers under psychological stress [108,203]. Neuroimaging studies have also confirmed that CBD administration decreased activity in limbic and paralimbic regions during emotional face processing tasks [175,178,204]. In Parkinson’s patients, CBD ameliorated the motor symptoms and also improved the quality of life of these patients [205]. CBD also decreases anxiety in socially anxious individuals [206,207]. In sum, based on the accumulated data so far, CBD seems to have great therapeutic potential without the significant adverse psychoactive effects associated with THC and this is likely due to its multiple mechanisms of action. Of note, the precursor of CBD, Cannabidiolic acid (CBDA) seems to be 100–1000 times more potent than CBD in reducing toxin-induced vomiting and nausea in animal models [208,209] and has also been shown to act as an anxiolytic agent by preventing stress-induced anxiogenic-like behavior [210].

10.3. Combinations of THC and CBD

Combinations of THC and CBD have also been tested for different illnesses, and the results indicate that preparations with lower THC and higher CBD ratios are usually more efficient than the inverse (higher THC and lower CBD ratios) [11,145]. For example, Nabiximols, a cannabis-based preparation containing THC and CBD in a ratio of 1:1, was approved in Canada for the relief of Multiple Sclerosis (MS) spasticity or cancer pain [168,169]. Interestingly, in preclinical tests ineffectively low doses of THC or CBDA when given alone turn into very effective treatment for acute nausea and vomiting when given in combination [168]. These very low doses of combined CBDA and THC also showed anti-inflammatory effects and reduced pain sensation in these preclinical studies, although CBDA by itself was also capable of such effects [168].

10.4. Synthetic Cannabinoids

Human studies using synthetic cannabinoids are rare in the context of patients with depression and anxiety [108,128]. Depression and anxiety in most of these studies were evaluated only as secondary analyses within healthy populations or populations suffering from another illness such as pain, cancer, and PTSD [5,108,128,178]. Nabilone (Cesamet) and dronabinol, synthetic analogues of THC, are two FDA cannabinoids approved and used to treat different ailments, including nausea and vomiting, associated with cancer chemotherapy and the treatment of anorexia associated with weight loss in patients with AIDS as well as multiple sclerosis (MS) in different countries [108]. In relation to depression and anxiety, nabilone improved the quality of life and reduced pain in a randomized, double-blind, placebo-controlled trial in patients with fibromyalgia [211], and reduced anxiety and pain in cancer patients when used as an adjunct to opioid treatment [212]. These data are in line with results obtained in studies using dronabinol, another synthetic THC, in patients with chronic central neuropathic pain or fibromyalgia [5,213,214]. Dronabinol has been studied in healthy populations in a small double-blind randomized controlled trial (RCT) (n = 16; 7.5 mg) which showed reduced limbic reactivity to angry or fearful faces during an emotional faces processing task [178,215]. Dronabinol was also efficient in fear extinction, a key component of PTSD and anxiety disorders [216,217,218].
Few research studies involving clinically anxious/depressive populations were analyzed [108], but it was concluded that nabilone significantly reduced anxiety [219,220] as it was seen to be associated with a 26.5% improvement in “generalized anxiety scores” [220]. This small study, however, involved patients also taking an antidepressant medication for mixed anxiety and mood disorders [178].

11. Discussion, Conclusions and Perspectives

Compelling evidence indicates an important role for the endocannabinoid system (ECS) in the regulation of mood disorders, including anxiety and depression. The involvement of the ECS in mood disorders is linked to its wide presence throughout different brain structures involved in the regulation of mood, emotion, and reward. In addition, there are strong scientific indications showing a wide range of interactions between the ECS and many other endogenous transmitter systems, with its capacity to modulate GABAergic and glutamatergic tones, as well as the release of dopamine, serotonin, opioids, norepinephrine, among many other effects, including regulating astrocyte and glial functions. Changes affecting ECS signaling produce a plethora of physiological and behavioral effects consistent with symptoms of depression and anxiety, in line with the hypothesis that a deficit in endocannabinoid signaling may yield a susceptibility to, or directly participate in, the development of a depressive episode [169]. Perturbations affecting the levels of eCBs seem to affect depression and anxiety symptomatology in humans and associated behaviors in animal models. In severe mood disorders, such as MDD and BD, decreases in eCBs and/or cannabinoid receptor signaling have been shown, whereas increasing the endocannabinoid tone seems to be beneficial in reducing depressive and anxiogenic behavior and symptoms. This has been shown by different approaches, including by preventing eCB degradation by inhibitors for FAAH and preventing eCB action by antagonism of CB1R. One clear example, mentioned above, is the distinct insight provided from CB1R blockade using rimonabant as an anti-obesity drug, which necessitated its removal from the market because of induction of severe symptoms of depression and suicidality. In addition, SNPs for two major players in the ECS, FAAH and CB1R, were linked to depression and anxiety.
Nevertheless, the field is still unclear when it comes to recommendations regarding the use of cannabinoids in treating depression, anxiety, and effects of stress, whether as antidepressants or as adjuncts to other antidepressants. Cannabis and its derivatives from both plant-based and synthetic origins are used in many circumstances to treat pain, nausea/vomiting, stress, anxiety and depression co-morbid with serious diseases such as cancer, MS, PTSD, Parkinson’s disease and other illnesses. Although showing a positive effect of cannabinoids in ameliorating the quality of life of many patients, including an improvement in their mood in general, there are many weaknesses and flaws surrounding the studies conducted so far. Different systematic and meta-analysis reviews agree on these aspects and list among the flaws detected: the small sample size of the studies; the lack of RCTs; the use of diverse preparations; different dosages and methods of administration; and the use of healthy individuals, with no regard for sex and age differences. In some circumstances, these weaknesses have led to conflicting and opposing conclusions [102,122]. This is greatly magnified in the contemplation of the use of cannabis or THC-enriched preparations for long-term medicinal use. The long-term use of cannabinoids, notably cannabis or THC-rich products, has significant deleterious effects, especially among humans with addictive liability patterns and can cause serious and permanent harm in adolescents. As mentioned above, between 50% and 95% of heavy users show cannabis withdrawal symptoms [140,141,142,143], with a recent meta-analysis reporting an overall prevalence of 47% among such users [144]. Chronic or heavy cannabis use has been shown to lead to cannabis use disorder (CUD), with strong links to other substance use disorders (SUD) and the symptoms of anhedonia and anxiety that are usually attached to them. In MDD and BD, the conclusions from the meta-analyses were clearly negative regarding the use of cannabis or THC-preparations over the long-term [108]. Although CBD is present in some of these preparations and may inhibit some side effects of THC, there is no certainty that CBD can abolish them all. Furthermore, during the last two decades, the potency of cannabis products has escalated together with a tendency to increase THC and decrease CBD concentrations [11,133,221]. This fact will certainly amplify the negative impact of cannabis self-medication, especially among the young population and patients already severely impacted by depression, anxiety and stress. Some authors of systematic reviews and meta-analyses have concluded that there are no clear benefits in using cannabis and certain cannabinoid preparations in combatting depression and anxiety in the long-term [108,109,128]. This has been accompanied by warnings that long-term cannabis or preparations high in THC may in fact worsen these disorders [108,128]. There is a definite need for more RCT studies, especially in severe mood disorders such as MDD and BD, where there is a lack of such studies. There is expectation of some progress with future studies that consider these pitfalls. Some of these studies are ongoing or at the stage of recruiting subjects [108]. In the interim, it is very difficult to draw a definitive conclusion in relation to the use of cannabinoids for treatment of depression and anxiety. Nevertheless, a common conclusion is that short-term judicious use of cannabis or derivatives lower in THC and higher in CBD may be beneficial in reducing anxiety and depression in different disorders. However, the long-term prospect of using such cannabis-based medicinal preparations may need much more careful scrutiny as it may result in the converse of the desired effect. In addition, the possibility of inducing cannabis use disorder or addiction to cannabis and cannabinoid preparations rich in THC is a distinct possibility that should not be overlooked among certain patients, especially since the prevalence of such disorders is high and rising [144]. CBD-enriched preparations appear to show a more consistent, beneficial effect in ameliorating mood under different conditions, which could present some promise for future use in mood-related disorders. Most studies from cannabis abusers have shown that high concentrations of THC and high ratios of THC:CBD were associated with more vigorous euphoria, but also with higher anxiety, depression and psychotic symptoms. It has also been shown that CBD attenuates some effects of THC such as anxiety, cognitive deficits, and psychosis in heavy cannabis users, and high CBD:THC ratios produce an effect opposite to the high THC:CBD concentrations [11,145,175,178,222,223,224,225,226]. However, the mechanisms by which CBD and THC interact or alleviate negative consequences are not clear, due in part to the multiple biological targets that both compounds can modulate. There is an undeniable need to identify the roles of these targets of THC and CBD, including different receptor complexes to clarify the different, and in some cases, opposite biological effects on the brain. This is particularly important with the view of identifying novel therapeutic targets for multiple illnesses, including anxiety and depression, using cannabinoid preparations devoid of side effects apparent during chronic treatment, including after the use of very low dosages [227], as well as to possibly reverse the adverse consequences of long-term cannabis use and abuse or withdrawal signs in CUD individuals [11,145].
In summary, there are credible arguments for a functional role of the ECS in mood disorders including anxiety and depression. There is also immense interest regarding clinical research in cannabinoids, in part due to the fact that cannabis was declared to be an illicit drug for a long time, hindering its legitimate use in medicine if proven to have benefit. In mood disorders, there is a need for multiple comprehensive RCTs, as the data so far are not completely convincing.

Author Contributions

A.H. and S.R.G. wrote the manuscript and B.K.M., A.H. and S.R.G. edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This project was funded by grants from NIDA DA042178 (to BKM and SRG), CIHR PJT-148633 (to SRG).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. ElSohly, M.A.; Slade, D. Chemical constituents of marijuana: The complex mixture of natural cannabinoids. Life Sci. 2005, 78, 539–548. [Google Scholar] [CrossRef]
  2. Gaoni, Y.; Mechoulam, R. Isolation, structure and partialsynthesis of anactive constituent of hashish. J. Am. Chem. Soc. 1964, 86, 1646–1647. [Google Scholar] [CrossRef]
  3. World Health Organization, Office on Drugs and Crime (UNDOC), Report 2022. Available online: https://www.unodc.org/unodc/en/press/releases/2022/June/unodc-world-drug-report-2022 (accessed on 20 July 2022).
  4. IsHak, W.W.; Wen, R.Y.; Naghdechi, L.; Vanle, B.; Dang, J.; Knosp, M.; Dascal, J.; Marcia, L.; Gohar, Y.; Louy, C.; et al. Pain and depression: A systematic review. Harv. Rev. Psychiatry 2018, 26, 352–363. [Google Scholar] [CrossRef]
  5. Huang, W.; Chen, W.; Zhang, X. Endocannabinoid system: Role in depression, reward and pain control. Mol. Med. Rep. 2016, 14, 2899–2903. [Google Scholar] [CrossRef]
  6. ElSohly, M.A.; Radwan, M.M.; Gul, W.; Chandra, S.; Galal, A. Phytochemistry of Cannabis sativa L. Prog. Chem. Org. Nat. Prod. 2017, 103, 1–36. [Google Scholar] [CrossRef]
  7. Adams, R.; Hunt, M.; Clark, J.H. Structure of cannabidiol, a product isolated from the marihuana extract of Minnesota wild hemp. I. J. Am. Chem. Soc. 1940, 62, 196–200. [Google Scholar] [CrossRef]
  8. Casadio, P.; Fernandes, C.; Murray, R.M.; Di Forti, M. Cannabis use in young people: The risk for schizophrenia. Neurosci. Biobehav. Rev. 2011, 35, 1779–1787. [Google Scholar] [CrossRef]
  9. Pacher, P.; Bátkai, S.; Kunos, G. The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacol. Rev. 2006, 58, 389–462. [Google Scholar] [CrossRef]
  10. Micale, V.; Di Marzo, V.; Sulcova, A.; Wotjak, C.T.; Drago, F. Endocannabinoid system and mood disorders: Priming a target for new therapies. Pharmacol. Ther. 2013, 138, 18–37. [Google Scholar] [CrossRef]
  11. Madras, B.K. Cannabinoid and Marijuana Neurobiology. In Cannabis in Medicine; Springer: Cham, Switzerland, 2020; pp. 25–47. [Google Scholar]
  12. Matsuda, L.A.; Lolait, S.J.; Brownstein, M.J.; Young, A.C.; Bonner, T.I. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 1990, 346, 561–564. [Google Scholar] [CrossRef]
  13. Munro, S.; Thomas, K.L.; Abu-Shaar, M. Molecular characterization of a peripheral receptor for cannabinoids. Nature 1993, 365, 61–65. [Google Scholar] [CrossRef]
  14. Di Marzo, V.; Bifulco, M.; De Petrocellis, L. The endocannabinoid system and its therapeutic exploitation. Nat. Rev. Drug Discov. 2004, 3, 771–784. [Google Scholar] [CrossRef]
  15. Pertwee, R.G.; Howlett, A.C.; Abood, M.E.; Alexander, S.P.H.; Di Marzo, V.; Elphick, M.R.; Greasley, P.J.; Hansen, H.S.; Kunos, G.; Mackie, K.; et al. International union of basic and clinical pharmacology. LXXIX. Cannabinoid receptors and their ligands: Beyond CB1 and CB2. Pharmacol. Rev. 2010, 62, 588–631. [Google Scholar] [CrossRef]
  16. Herkenham, M.; Lynn, A.B.; Little, M.D.; Johnson, M.R.; Melvin, L.S.; de Costa, B.R.; Rice, K.C. Cannabinoid receptor localization in brain. Proc. Natl. Acad. Sci. USA 1990, 87, 1932–1936. [Google Scholar] [CrossRef]
  17. Herkenham, M.; Lynn, A.B.; de Costa, B.R.; Richfield, E.K. Neuronal localization of cannabinoid receptors in the basal ganglia of the rat. Brain Res. 1991, 547, 267–274. [Google Scholar] [CrossRef]
  18. Hohmann, A.G.; Herkenham, M. Localization of cannabinoid CB(1) receptor mRNA in neuronal subpopulations of rat striatum: A double-label in situ hybridization study. Synapse 2000, 7, 71–80. [Google Scholar] [CrossRef]
  19. Sun, Y.; Alexander, S.P.; Kendall, D.A.; Bennett, A.J. Cannabinoids and PPARalpha signalling. Biochem. Soc. Trans. 2006, 34, 1095–1097. [Google Scholar] [CrossRef]
  20. Sawzdargo, M.; Nguyen, T.; Lee, D.K.; Lynch, K.R.; Cheng, R.; Heng, H.H.; George, S.R.; O’Dowd, B.F. Identification and cloning of three novel human G protein-coupled receptor genes GPR52, PsiGPR53 and GPR55: GPR55 is extensively expressed in human brain. Brain Res. Mol. Brain Res. 1999, 64, 193–198. [Google Scholar] [CrossRef]
  21. Shi, Q.X.; Yang, L.K.; Shi, W.L.; Wang, L.; Zhou, S.-M.; Guan, S.-Y.; Zhao, M.-G.; Yang, Q. The novel cannabinoid receptor GPR55 mediates anxiolytic-like effects in the medial orbital cortex of mice with acute stress. Mol. Brain 2017, 10, 38. [Google Scholar] [CrossRef]
  22. Ryberg, E.; Larsson, N.; Sjögren, S.; Hjorth, S.; Hermansson, N.O.; Leonova, J.; Elebring, T.; Nilsson, K.; Drmota, T.; Greasley, P.J. The orphan receptor GPR55 is a novel cannabinoid receptor. Br. J. Pharmacol. 2007, 152, 1092–1101. [Google Scholar] [CrossRef]
  23. Overton, H.A.; Babbs, A.J.; Doel, S.M.; Fyfe, M.C.; Gardner, L.S.; Griffin, G.; Jackson, H.C.; Procter, M.J.; Rasamison, C.M.; Tang-Christensen, M.; et al. Deorphanization of a G protein-coupled receptor for oleoylethanolamide and its use in the discovery of small-molecule hypophagic agents. Cell Metab. 2006, 3, 167–175. [Google Scholar] [CrossRef]
  24. Morales, P.; Hurst, D.P.; Reggio, P.H. Molecular targets of the phytocannabinoids: A complex picture. Prog. Chem. Org. Nat. Prod. 2017, 103, 103–131. [Google Scholar]
  25. Pérez-Olives, C.; Rivas-Santisteban, R.; Lillo, J.; Navarro, G.; Franco, R. Recent Advances in the Potential of Cannabinoids for Neuroprotection in Alzheimer’s, Parkinson’s, and Huntington’s Diseases. Adv. Exp. Med. Biol. 2021, 1264, 81–92. [Google Scholar] [CrossRef]
  26. Callén, L.; Moreno, E.; Barroso-Chinea, P.; Moreno-Delgado, D.; Cortés, A.; Mallol, J.; Casadó, V.; Lanciego, J.L.; Franco, R.; Lluis, C.; et al. Cannabinoid receptors CB1 and CB2 form functional heteromers in brain. J. Biol. Chem. 2012, 287, 20851–20865. [Google Scholar] [CrossRef]
  27. Glass, M.; Felder, C.C. Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors augments cAMP accumulation in striatal neurons: Evidence for a Gs linkage to the CB1 receptor. J. Neurosci. 1997, 17, 5327–5333. [Google Scholar] [CrossRef]
  28. Przybyla, J.A.; Watts, V.J. Ligand-induced regulation and localization of cannabinoid CB1 and dopamine D2L receptor heterodimers. J. Pharmacol. Exp. Ther. 2010, 332, 710–719. [Google Scholar] [CrossRef]
  29. Pazos, M.R.; Mohammed, N.; Lafuente, H.; Santos, M.; Martínez-Pinilla, E.; Moreno, E.; Valdizan, E.; Romero, J.; Pazos, A.; Franco, R.; et al. Mechanisms of cannabidiol neuroprotection in hypoxic-ischemic newborn pigs: Role of 5HT(1A) and CB2 receptors. Neuropharmacology 2013, 71, 282–291. [Google Scholar] [CrossRef]
  30. Vinals, X.; Moreno, E.; Lanfumey, L.; Cordomí, A.; Pastor, A.; De La Torre, R.; Gasperini, P.; Navarro, G.; Howell, L.A.; Pardo, L.; et al. Cognitive impairment induced by Delta9-tetrahydrocannabinol occurs through heteromers between cannabinoid CB1 and serotonin 5-HT2A receptors. PLoS Biol. 2015, 13, e1002194. [Google Scholar] [CrossRef]
  31. Orgado, J.M.; Fernández-Ruiz, J.; Romero, J. The endocannabinoid system in neuropathological states. Int. Rev. Psychiatry 2009, 21, 172–180. [Google Scholar] [CrossRef]
  32. Martınez-Pinilla, E.; Reyes-Resina, I.; Oñatibia-Astibia, A.; Zamarbide, M.; Ricobaraza, A.; Navarro, G.; Moreno, E.; Dopeso-Reyes, I.; Sierra, S.; Rico, A.; et al. CB1 and GPR55 receptors are co-expressed and form heteromers in rat and monkey striatum. Exp. Neurol. 2014, 261, 44–52. [Google Scholar] [CrossRef]
  33. Devane, W.A.; Hanus, L.; Breuer, A.; Pertwee, R.G.; Stevenson, L.A.; Griffin, G.; Gibson, D.; Mandelbaum, A.; Etinger, A.; Mechoulam, R. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 1992, 258, 1946–1949. [Google Scholar] [CrossRef]
  34. Mechoulam, R.; Ben-Shabat, S.; Hanus, L.; Ligumsky, M.; Kaminski, N.E.; Schatz, A.R.; Gopher, A.; Almog, S.; Martin, B.R.; Compton, D.R.; et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem. Pharmacol. 1995, 50, 83–90. [Google Scholar] [CrossRef]
  35. Sugiura, T.; Kondo, S.; Sukagawa, A.; Nakane, S.; Shinoda, A.; Itoh, K.; Yamashita, A.; Waku, K. 2-Arachidonoylglycerol: A possible endogenous cannabinoid receptor ligand in brain. Biochem. Biophys. Res. Commun. 1995, 215, 89–97. [Google Scholar] [CrossRef]
  36. Mechoulam, R.; Hanus, L.O.; Pertwee, R.; Howlett, A.C. Early phytocannabinoid chemistry to endocannabinoids and beyond. Nat. Rev. Neurosci. 2014, 15, 757–764. [Google Scholar] [CrossRef]
  37. Hanus, L.; Abu-Lafi, S.; Fride, E.; Breuer, A.; Vogel, Z.; Shalev, D.E.; Kustanovich, I.; Mechoulam, R. 2-arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc. Natl. Acad. Sci. USA 2001, 98, 3662–3665. [Google Scholar] [CrossRef]
  38. Huang, S.M.; Bisogno, T.; Trevisani, M.; Al-Hayani, A.; De Petrocellis, L.; Fezza, F.; Tognetto, M.; Petros, T.J.; Krey, J.F.; Chu, C.J.; et al. An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors. Proc. Natl. Acad. Sci. USA 2002, 99, 8400–8405. [Google Scholar] [CrossRef]
  39. Porter, A.C.; Sauer, J.M.; Knierman, M.D.; Becker, G.W.; Berna, M.J.; Bao, J.; Nomikos, G.G.; Carter, P.; Bymaster, F.P.; Leese, A.B.; et al. Characterization of a novel endocannabinoid, virodhamine, with antagonist activity at the CB1 receptor. J. Pharmacol. Exp. Ther. 2002, 301, 1020–1024. [Google Scholar] [CrossRef]
  40. Leggett, J.D.; Aspley, S.; Beckett, S.R.; D’Antona, A.M.; Kendall, D.A.; Kendall, D.A. Oleamide is a selective endogenous agonist of rat and human CB1 cannabinoid receptors. Br. J. Pharmacol. 2004, 141, 253–262. [Google Scholar] [CrossRef]
  41. Cravatt, B.F.; Giang, D.K.; Mayfield, S.P.; Boger, D.L.; Lerner, R.A.; Gilula, N.B. Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature 1996, 384, 83–87. [Google Scholar] [CrossRef]
  42. Blankman, J.L.; Simon, G.M.; Cravatt, B.F. A comprehensive profile of brain enzymes that hydrolyze the endocannabinoid 2-arachidonoylglycerol. Chem. Biol. 2007, 14, 1347–1356. [Google Scholar] [CrossRef]
  43. Kozak, K.R.; Marnett, L.J. Oxidative metabolism of endocannabinoids. Prostaglandins Leukot. Essent. Fat. Acids (PLEFA) 2002, 66, 211–220. [Google Scholar] [CrossRef]
  44. van der Stelt, M.; van Kuik, J.A.; Bari, M.; van Zadelhoff, G.; Leeflang, B.R.; Veldink, G.A.; Finazzi-Agrò, A.; Vliegenthart, J.F.; Maccarrone, M. Oxygenated metabolites of anandamide and 2-arachidonoylglycerol: Conformational analysis and interaction with cannabinoid receptors, membrane transporter, and fatty acid amide hydrolase. J. Med. Chem. 2002, 45, 3709–3720. [Google Scholar] [CrossRef]
  45. Woodward, D.F.; Carling, R.W.; Cornell, C.L.; Fliri, H.G.; Martos, J.L.; Pettit, S.N.; Liang, Y.; Wang, J.W. The pharmacology and therapeutic relevance of endocannabinoid derived cyclo-oxygenase (COX)-2 products. Pharmacol. Ther. 2008, 120, 71–80. [Google Scholar] [CrossRef]
  46. Wilson, R.I.; Nicoll, R.A. Endogenous cannabinoids mediate retrograde signalling at hippocampal synapses. Nature 2001, 410, 588–592, Erratum in: Nature 2001, 411, 974. [Google Scholar] [CrossRef]
  47. Araque, A.; Castillo, P.E.; Manzoni, O.J.; Tonini, R. Synaptic functions of endocannabinoid signaling in health and disease. Neuropharmacology 2017, 124, 13–24. [Google Scholar] [CrossRef]
  48. Castillo, P.E.; Younts, T.J.; Chávez, A.E.; Hashimotodani, Y. Endocannabinoid signaling and synaptic function. Neuron 2012, 76, 70–81. [Google Scholar] [CrossRef]
  49. Kano, M.; Ohno-Shosaku, T.; Hashimotodani, Y.; Uchigashima, M.; Watanabe, M. Endocannabinoid-mediated control of synaptic transmission. Physiol. Rev. 2009, 89, 309–380. [Google Scholar] [CrossRef]
  50. Metna-Laurent, M.; Marsicano, G. Rising stars: Modulation of brain functions by astroglial type-1 cannabinoid receptors. Glia 2015, 63, 353–364. [Google Scholar] [CrossRef]
  51. Navarrete, M.; Díez, A.; Araque, A. Astrocytes in endocannabinoid signalling. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130599. [Google Scholar] [CrossRef]
  52. Chevaleyre, V.; Takahashi, K.A.; Castillo, P.E. Endocannabinoid-mediated synaptic plasticity in the CNS. Annu. Rev. Neurosci. 2006, 29, 37–76. [Google Scholar] [CrossRef]
  53. Mátyás, F.; Gabriella, M.U.; Masahiko, W.; Ken, M.; Andreas, Z.; Tamás, F.F.; István, K. Identification of the sites of 2-arachidonoylglycerol synthesis and action imply retrograde endocannabinoid signaling at both GABAergic and glutamatergic synapses in the ventral tegmental area. Neuropharmacology 2008, 54, 95–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Fitzgerald, M.L.; Shobin, E.; Pickel, V.M. Cannabinoid modulation of the dopaminergic circuitry: Implications for limbic and striatal output. Prog. Neuropsychopharmacol. Biol. Psychiatry 2012, 38, 21–29. [Google Scholar] [CrossRef] [PubMed]
  55. Covey, D.P.; Mateo, Y.; Sulzer, D.; Cheer, J.F.; Lovinger, D.M. Endocannabinoid modulation of dopamine neurotransmission. Neuropharmacology 2017, 124, 52–61. [Google Scholar] [CrossRef] [PubMed]
  56. Szabo, B.; Siemes, S.; Wallmichrath, I. Inhibition of GABAergic neurotransmission in the ventral tegmental area by cannabinoids. Eur. J. Neurosci. 2002, 15, 2057–2061. [Google Scholar] [CrossRef] [PubMed]
  57. Riegel, A.C.; Lupica, C.R. Independent presynaptic and postsynaptic mechanisms regulate endocannabinoid signaling at multiple synapses in the ventral tegmental area. J. Neurosci. 2004, 24, 11070–11078. [Google Scholar] [CrossRef] [PubMed]
  58. Melis, M.; Pistis, M. Hub and switches: Endocannabinoid signalling in midbrain dopamine neurons. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2012, 367, 3276–3285. [Google Scholar] [CrossRef]
  59. Wang, T.; Collet, J.P.; Shapiro, S.; Ware, M.A. Adverse effects of medical cannabinoids: A systematic review. CMAJ 2008, 178, 1669–1678. [Google Scholar] [CrossRef]
  60. Talani, G.; Licheri, V.; Biggio, F.; Locci, V.; Mostallino, M.C.; Secci, P.P.; Melis, V.; Dazzi, L.; Carta, G.; Banni, S.; et al. Enhanced Glutamatergic Synaptic Plasticity in the Hippocampal CA1 Field of Food-Restricted Rats: Involvement of CB1 Receptors. Neuropsychopharmacology 2016, 41, 1308–1318. [Google Scholar] [CrossRef]
  61. Melis, M.; Pistis, M.; Perra, S.; Muntoni, A.L.; Pillolla, G.; Gessa, G.L. Endocannabinoids mediate presynaptic inhibition of glutamatergic transmission in rat ventral tegmental area dopamine neurons through activation of CB1 receptors. J. Neurosci. 2004, 24, 53–62. [Google Scholar] [CrossRef]
  62. Spanagel, R. Cannabinoids and the endocannabinoid system in reward processing and addiction: From mechanisms to interventions. Dialogues Clin. Neurosci. 2020, 22, 241–250. [Google Scholar] [CrossRef]
  63. Robbe, D.; Kopf, M.; Remaury, A.; Bockaert, J.; Manzoni, O.J. Endogenous cannabinoids mediate long-term synaptic depression in the nucleus accumbens. Proc. Natl. Acad. Sci. USA 2002, 99, 8384–8388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zlebnik, N.E.; Cheer, J.F. Beyond the CB1 receptor: Is cannabidiol the answer for disorders of motivation? Annu. Rev. Neurosci. 2016, 39, 1–17. [Google Scholar] [CrossRef]
  65. Heifets, B.D.; Castillo, P.E. Endocannabinoid Signaling and Long-Term Synaptic Plasticity. Annu. Rev. Physiol. 2009, 71, 283–306. [Google Scholar] [CrossRef] [PubMed]
  66. Fowler, C.J. Anandamide uptake explained? Trends Pharmacol. Sci. 2012, 33, 181–185. [Google Scholar] [CrossRef] [PubMed]
  67. Poleszak, E.; Wośko, S.; Sławińska, K.; Szopa, A.; Wróbel, A.; Serefko, A. Cannabinoids in depressive disorders. Life Sci. 2018, 213, 18–24. [Google Scholar] [CrossRef]
  68. Hill, M.N.; Gorzalka, B.B. Is there a role for the endocannabinoid system in the etiology and treatment of melancholic depression? Behav. Pharmacol. 2005, 16, 333–352. [Google Scholar] [CrossRef] [PubMed]
  69. El-Alfy, A.T.; Ivey, K.; Robinson, K.; Ahmed, S.; Radwan, M.; Slade, D.; Khan, I.; ElSohly, M.; Ross, S. Antidepressant-like effect of Δ9-tetrahydrocannabinol and other cannabinoids isolated from Cannabis sativa L. Pharmacol. Biochem. Behav. 2010, 95, 434–442. [Google Scholar] [CrossRef]
  70. Bambico, F.R.; Hattan, P.R.; Garant, J.P.; Gobbi, G. Effect of delta-9-tetrahydrocannabinol on behavioral despair and on pre- and postsynaptic serotonergic transmission. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2012, 38, 88–96. [Google Scholar] [CrossRef] [PubMed]
  71. Bambico, F.R.; Duranti, A.; Tontini, A.; Tarzia, G.; Gobbi, G. Endocannabinoids in the treatment of mood disorders: Evidence from animal models. Curr. Pharm. Des. 2009, 15, 1623–1646. [Google Scholar] [CrossRef] [PubMed]
  72. Adamczyk, P.; Golda, A.; McCreary, A.C.; Filip, M.; Przegaliriski, E. Activation of endocannabinoid transmission induces antidepressant-like effects in rats. Acta Physiol. Pol. 2008, 59, 217. [Google Scholar]
  73. Hayase, T. Differential effects of TRPV1 receptor ligands against nicotine-induced depression-like behaviors. BMC Pharmacol. 2011, 11, 6. [Google Scholar] [CrossRef]
  74. Umathe, S.N.; Manna, S.S.; Jain, N.S. Involvement of endocannabinoids in antidepressant and anti-compulsive effect of fluoxetine in mice. Behav. Brain Res. 2011, 223, 125–134. [Google Scholar] [CrossRef]
  75. Rubino, T.; Parolaro, D. Long lasting consequences of cannabis exposure in adolescence. Mol. Cell. Endocrinol. 2008, 286, S108–S113. [Google Scholar] [CrossRef] [PubMed]
  76. Long, J.Z.; Li, W.; Booker, L.; Burston, J.J.; Kinsey, S.G.; Schlosburg, J.E.; Pavon, F.J.; Serrano, A.M.; Selley, D.E.; Parsons, L.H.; et al. Selective blockade of 2-arachidonoylglycerol hydrolysis produces cannabinoid behavioral effects. Nat. Chem. Biol. 2009, 5, 37–44. [Google Scholar] [CrossRef]
  77. O’Brien, L.D.; Wills, K.L.; Segsworth, B.; Dashney, B.; Rock, E.M.; Limebeer, C.L.; Parker, L.A. Effect of chronic exposure to rimonabant and phytocannabinoids on anxiety-like behavior and saccharin palatability. Pharmacol. Biochem. Behav. 2013, 103, 597–602. [Google Scholar] [CrossRef] [PubMed]
  78. Patel, R.; Wilson, R.; Jackson, R.; Ball, M.; Shetty, H.; Broadbent, M.; Stewart, R.; McGuire, P.; Bhattacharyya, S. Cannabis use and treatment resistance in first episode psychosis: A natural language processing study. Lancet 2015, 385, S79. [Google Scholar] [CrossRef] [PubMed]
  79. Moreira, F.A.; Kaiser, N.; Monory, K.; Lutz, B. Reduced anxietylike behaviour induced by genetic and pharmacological inhibition of the endocannabinoid-degrading enzyme fatty acid amide hydrolase (FAAH) is mediated by CB1 receptors. Neuropharmacology 2008, 54, 141–150. [Google Scholar] [CrossRef]
  80. Patel, S.; Hillard, C.J. Role of endocannabinoid signaling in anxiety and depression. Curr. Top. Behav. Neurosci. 2009, 1, 347–371. [Google Scholar]
  81. Haller, J.; Varga, B.; Ledent, C.; Freund, T.F. CB1 cannabinoid receptors mediate anxiolytic effects: Convergent genetic and pharmacological evidence with CB1-specific agents. Behav. Pharmacol. 2004, 15, 299–304. [Google Scholar] [CrossRef]
  82. Spanagel, R.; Bilbao, A. Approved cannabinoids for medical purposes—Comparative systematic review and meta-analysis for sleep and appetite. Neuropharmacology 2021, 196, 108680. [Google Scholar] [CrossRef]
  83. Degroot, A.; Nomikos, G.G. Genetic deletion and pharmacological blockade of CB1 receptors modulates anxiety in the shock-probe burying test. Eur. J. Neurosci. 2004, 20, 1059–1064. [Google Scholar] [CrossRef]
  84. Kathuria, S.; Gaetani, S.; Fegley, D.; Valiño, F.; Duranti, A.; Tontini, A.; Mor, M.; Tarzia, G.; La Rana, G.; Calignano, A.; et al. Modulation of anxiety through blockade of anandamide hydrolysis. Nat. Med. 2003, 9, 76–81. [Google Scholar] [CrossRef]
  85. Bortolato, M.; Campolongo, P.; Mangieri, R.A.; Scattoni, M.L.; Frau, R.; Trezza, V.; La Rana, G.; Russo, R.; Calignano, A.; Gessa, G.L.; et al. Anxiolytic-like properties of the anandamide transport inhibitor AM404. Neuropsychopharmacology 2006, 31, 2652–2659. [Google Scholar] [CrossRef] [Green Version]
  86. Gorzalka, B.B.; Hill, M.N. Putative role of endocannabinoid signaling in the etiology of depression and actions of antidepressants. Prog. Neuropsychopharmacol. Biol. Psychiatry 2011, 35, 1575–1585. [Google Scholar] [CrossRef]
  87. Shearman, L.P.; Rosko, K.M.; Fleischer, R.; Wang, J.; Xu, S.; Tong, X.S.; Rocha, B.A. Antidepressant-like and anorectic effects of the cannabinoid CB1 receptor inverse agonist AM251 in mice. Behav. Pharmacol. 2003, 14, 573–582. [Google Scholar] [CrossRef] [PubMed]
  88. Tzavara, E.T.; Davis, R.J.; Perry, K.W.; Li, X.; Salhoff, C.; Bymaster, F.P.; Witkin, J.M.; Nomikos, G.G. The CB1 receptor antagonist selectively increases monoaminergic neurotransmission in the medial prefrontal cortex: Implications for therapeutic actions. Br. J. Pharmacol. 2003, 138, 544–553. [Google Scholar] [CrossRef]
  89. Witkin, J.M.; Tzavara, E.T.; Davis, R.J.; Li, X.; Nomikos, G.G. A therapeutic role for cannabinoid CB1 receptor antagonists in major depressive disorders. Trends Pharmacol. Sci. 2005, 26, 609–617. [Google Scholar] [CrossRef] [PubMed]
  90. Griebel, G.; Stemmelin, J.; Scatton, B. Effects of the cannabinoid CB1 receptor antagonist rimonabant in models of emotional reactivity in rodents. Biol. Psychiatry 2005, 57, 261–267. [Google Scholar] [CrossRef] [PubMed]
  91. Sanchis-Segura, C.; Cline, B.H.; Marsicano, G.; Lutz, B.; Spanagel, R. Reduced sensitivity to reward in CB1 knockout mice. Psychopharmacology 2004, 176, 223–232. [Google Scholar] [CrossRef]
  92. Haller, J.; Varga, B.; Ledent, C.; Barna, I.; Freund, T.F. Context-dependent effects of CB1 cannabinoid gene disruption on anxiety-like and social behaviour in mice. Eur. J. Neurosci. 2004, 19, 1906–1912. [Google Scholar] [CrossRef]
  93. Haller, J.; Bakos, N.; Szirmay, M.; Ledent, C.; Freund, T.F. The effects of genetic and pharmacological blockade of the CB1 cannabinoid receptor on anxiety. Eur. J. Neurosci. 2002, 16, 1395–1398. [Google Scholar] [CrossRef]
  94. Mikics, E.; Vas, J.; Aliczki, M.; Halasz, J.; Haller, J. Interactions between the anxiogenic effects of CB1 gene disruption and 5-HT3 neurotransmission. Behav. Pharmacol. 2009, 20, 265–272. [Google Scholar] [CrossRef]
  95. Aso, E.; Ozaita, A.; Valdizán, E.M.; Ledent, C.; Pazos, Á.; Maldonado, R.; Valverde, O. BDNF impairment in the hippocampus is related to enhanced despair behavior in CB1 knockout mice. J. Neurochem. 2008, 105, 565–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Steiner, M.A.; Wanisch, K.; Monory, K.; Marsicano, G.; Borroni, E.; Bächli, H.; Holsboer, F.; Lutz, B.; Wotjak, C.T. Impaired cannabinoid receptor type 1 signaling interferes with stress-coping behavior in mice. Pharm. J. 2008, 8, 196–208. [Google Scholar] [CrossRef]
  97. Patel, S.; Hillard, C.J. Pharmacological evaluation of cannabinoid receptor ligands in a mouse model of anxiety: Further evidence for an anxiolytic role for endogenous cannabinoid signaling. J. Pharmacol. Exp. Ther. 2006, 318, 304–311. [Google Scholar] [CrossRef] [PubMed]
  98. Hill, M.N.; Carrier, E.J.; McLaughlin, R.J.; Morrish, A.C.; Meier, S.E.; Hillard, C.J.; Gorzalka, B.B. Regional alterations in the endocannabinoid system in an animal model of depression: Effects of concurrent antidepressant treatment. J. Neurochem. 2008, 106, 2322–2336. [Google Scholar] [CrossRef] [PubMed]
  99. Hill, M.N.; Gorzalka, B.B. Enhancement of anxiety-like responsiveness to the cannabinoid CB(1) receptor agonist HU-210 following chronic stress. Eur. J. Pharmacol. 2004, 499, 291–295. [Google Scholar] [CrossRef] [PubMed]
  100. Reich, C.G.; Taylor, M.E.; McCarthy, M.M. Differential effects of chronic unpredictable stress on hippocampal CB1 receptors in male and female rats. Behav. Brain Res. 2009, 203, 264–269. [Google Scholar] [CrossRef]
  101. Hill, K.P. Medical marijuana for treatment of chronic pain and other medical and psychiatric problems: A clinical review. JAMA 2015, 313, 2474–2483. [Google Scholar] [CrossRef] [PubMed]
  102. Rademacher, D.J.; Meier, S.E.; Shi, L.; Ho WS, V.; Jarrahian, A.; Hillard, C.J. Effects of acute and repeated restraint stress on endocannabinoid content in the amygdala, ventral striatum, and medial prefrontal cortex in mice. Neuropharmacology 2008, 54, 108–116. [Google Scholar] [CrossRef]
  103. Rossi, S.; De Chiara, V.; Musella, A.; Kusayanagi, H.; Mataluni, G.; Bernardi, G.; Usiello, A.; Centonze, D. Chronic psychoemotional stress impairs cannabinoid-receptor-mediated control of GABA transmission in the striatum. J. Neurosci. 2008, 28, 7284–7292. [Google Scholar] [CrossRef] [PubMed]
  104. Wamsteeker, J.I.; Kuzmiski, J.B.; Bains, J.S. Repeated stress impairs endocannabinoid signaling in the paraventricular nucleus of the hypothalamus. J. Neurosci. 2010, 30, 11188–11196. [Google Scholar] [CrossRef] [PubMed]
  105. Legare, C.A.; Raup-Konsavage, W.M.; Vrana, K.E. Therapeutic Potential of Cannabis, Cannabidiol, and Cannabinoid-Based Pharmaceuticals. Pharmacology 2022, 107, 131–149. [Google Scholar] [CrossRef] [PubMed]
  106. Gobbi, G.; Bambico, F.; Mangieri, R.; Bortolato, M.; Campolongo, P.; Solinas, M.; Cassano, T.; Morgese, M.G.; Debonnel, G.; Duranti, A.; et al. Antidepressant-like activity and modulation of brain monoaminergic transmission by blockade of anandamide hydrolysis. Proc. Natl. Acad. Sci. USA 2005, 102, 18620–18625. [Google Scholar] [CrossRef] [PubMed]
  107. Naidu, P.S.; Varvel, S.A.; Ahn, K.; Cravatt, B.F.; Martin, B.R.; Lichtman, A.H. Evaluation of fatty acid amide hydrolase inhibition in murine models of emotionality. Psychopharmacology 2007, 192, 61–70. [Google Scholar] [CrossRef]
  108. Chadwick, V.L.; Rohleder, C.; Koethe, D.; Leweke, F.M. Cannabinoids and the endocannabinoid system in anxiety, depression, and dysregulation of emotion in humans. Curr. Opin. Psychiatry 2020, 33, 20–42. [Google Scholar] [CrossRef] [PubMed]
  109. Black, N.; Stockings, E.; Campbell, G.; Tran, L.T.; Zagic, D.; Hall, W.D.; Farrell, M.; Degenhardt, L. Cannabinoids for the treatment of mental disorders and symptoms of mental disorders: A systematic review and meta-analysis. Lancet Psychiatry 2019, 6, 995–1010, Erratum in: Lancet Psychiatry 2020, 7, e3. [Google Scholar] [CrossRef]
  110. Hungund, B.L.; Vinod, K.Y.; Kassir, S.A.; Basavarajappa, B.S.; Yalamanchili, R.; Cooper, T.B.; Mann, J.J.; Arango, V. Upregulation of CB 1 receptors and agonist-stimulated [35 S] GTP γ S binding in the prefrontal cortex of depressed suicide victims. Mol. Psychiatry 2004, 9, 184–190. [Google Scholar] [CrossRef]
  111. Vinod, K.Y.; Xie, S.; Psychoyos, D.; Hungund, B.L.; Cooper, T.B.; Tejani-Butt, S.M. Dysfunction in fatty acid amide hydrolase is associated with depressive-like behavior in Wistar Kyoto rats. PLoS ONE 2012, 7, e36743. [Google Scholar] [CrossRef]
  112. Mato, S.; Pilar-Cuéllar, F.; Valdizán, E.M.; González-Maeso, J.; Rodríguez-Puertas, R.; Meana, J.; Sallés, J.; Crespo-Facorro, B.; Pazos, Á. Selective up-regulation of cannabinoid CB1 receptor coupling to Go-proteins in suicide victims with mood disorders. Biochem. Pharmacol. 2018, 157, 258–265. [Google Scholar] [CrossRef]
  113. Koethe, D.; Llenos, I.C.; Dulay, J.R.; Hoyer, C.; Torrey, E.F.; Leweke, F.M.; Weis, S. Expression of CB1 cannabinoid receptor in the anterior cingulate cortex in schizophrenia, bipolar disorder, and major depression. J. Neural Transm. Vienna 2007, 114, 1055–1063. [Google Scholar] [CrossRef]
  114. Dlugos, A.; Childs, E.; Stuhr, K.L.; Hillard, C.J.; de Wit, H. Acute stress increases circulating anandamide and other N-acylethanolamines in healthy humans. Neuropsychopharmacology 2012, 37, 2416–2427. [Google Scholar] [CrossRef] [PubMed]
  115. Crombie, K.M.; Leitzelar, B.N.; Brellenthin, A.G.; Hillard, C.J.; Koltyn, K.F. Loss of exercise- and stress-induced increases in circulating 2-arachidonoylglycerol concentrations in adults with chronic PTSD. Biol. Psychol. 2019, 145, 1–7. [Google Scholar] [CrossRef] [PubMed]
  116. Kranaster, L.; Hoyer, C.; Aksay, S.S.; Bumb, J.M.; Müller, N.; Zill, P.; Schwarz, M.J.; Moll, N.; Lutz, B.; Bindila, L.; et al. Biomarkers for antidepressant efficacy of electroconvulsive therapy: An exploratory cerebrospinal fluid study. Neuropsychobiology 2019, 77, 13–22. [Google Scholar] [CrossRef] [PubMed]
  117. Mayo, L.M.; Asratian, A.; Lindé, J.; Holm, L.; Nätt, D.; Augier, G.; Stensson, N.; Vecchiarelli, H.A.; Balsevich, G.; Aukema, R.J.; et al. Protective effects of elevated anandamide on stress and fear-related behaviors: Translational evidence from humans and mice. Mol. Psychiatry 2020, 25, 993–1005. [Google Scholar] [CrossRef] [PubMed]
  118. Monteleone, P.; Bifulco, M.; Maina, G.; Tortorella, A.; Gazzerro, P.; Proto, M.C.; Di Filippo, C.; Monteleone, F.; Canestrelli, B.; Buonerba, G. Investigation of CNR1 and FAAH endocannabinoid gene polymorphisms in bipolar disorder and major depression. Pharmacol. Res. 2010, 61, 400–404. [Google Scholar] [CrossRef] [PubMed]
  119. Hill, M.N.; Gorzalka, B.B. Impairments in endocannabinoid signaling and depressive illness. JAMA 2009, 301, 1165–1166. [Google Scholar] [CrossRef]
  120. Hill, M.N.; Gorzalka, B.B. The endocannabinoid system and the treatment of mood and anxiety disorders. CNS Neurol. Disord. Drug Targets 2009, 8, 451–458. [Google Scholar] [CrossRef] [PubMed]
  121. Christensen, R.; Kristensen, P.K.; Bartels, E.M.; Bliddal, H.; Astrup, A. Efficacy and safety of the weight-loss drug rimonabant: A meta-analysis of randomised trials. Lancet 2007, 370, 1706–1713. [Google Scholar] [CrossRef]
  122. Nissen, S.E.; Nicholls, S.J.; Wolski, K.; Rodés-Cabau, J.; Cannon, C.P.; Deanfield, J.E.; Després, J.-P.; Kastelein, J.J.P.; Steinhubl, S.R.; Kapadia, S.; et al. Effect of rimonabant on progression of atherosclerosis in patients with abdominal obesity and coronary artery disease: The STRADIVARIUS randomized controlled trial. JAMA 2008, 299, 1547–1560. [Google Scholar] [CrossRef] [PubMed]
  123. Juhasz, G.; Chase, D.; Pegg, E.; Downey, D.; Toth, Z.G.; Stones, K.; Platt, H.; Mekli, K.; Payton, A.; Elliott, R.; et al. CNR1 gene is associated with high neuroticism and low agreeableness and interacts with recent negative life events to predict current depressive symptoms. Neuropsychopharmacology 2009, 34, 2019–2027. [Google Scholar] [CrossRef] [PubMed]
  124. Domschke, K.; Dannlowski, U.; Ohrmann, P.; Lawford, B.; Bauer, J.; Kugel, H.; Heindel, W.; Young, R.; Morris, P.; Arolt, V.; et al. Cannabinoid receptor 1 (CNR1) gene: Impact on antidepressant treatment response and emotion processing in major depression. Eur. Neuropsychopharmacol. 2008, 18, 751–759. [Google Scholar] [CrossRef] [PubMed]
  125. Moore, C.; Rana, S.; Coulter, C. Simultaneous identification of 2-carboxy-tetrahydrocannabinol, tetrahydrocannabinol, cannabinol and cannabidiol in oral fluid. J. Chromatogr. B 2007, 852, 459–464. [Google Scholar] [CrossRef] [PubMed]
  126. Volkow, N.D.; Baler, R.D.; Compton, W.M.; Weiss, S.R. Adverse health effects of marijuana use. N. Engl. J. Med. 2014, 370, 2219–2227. [Google Scholar] [CrossRef] [PubMed]
  127. Volkow, N.D.; Swanson, J.M.; Evins, A.E.; DeLisi, L.E.; Meier, M.H.; Gonzalez, R.; Bloomfield, M.A.; Curran, H.V.; Baler, R. Effects of Cannabis Use on Human Behavior, Including Cognition, Motivation, and Psychosis: A Review. JAMA Psychiatry 2016, 73, 292–297. [Google Scholar] [CrossRef] [PubMed]
  128. Lowe, D.J.E.; Sasiadek, J.D.; Coles, A.S.; George, T.P. Cannabis and mental illness: A review. Eur. Arch. Psychiatry Clin. Neurosci. 2019, 269, 107–120. [Google Scholar] [CrossRef] [PubMed]
  129. Solowij, N.; Stephens, R.S.; Roffman, R.A.; Babor, T.; Kadden, R.; Miller, M.; Christiansen, K.; McRee, B.; Vendetti, J. Cognitive functioning of long-term heavy cannabis users seeking treatment. JAMA 2002, 287, 1123–1131. [Google Scholar] [CrossRef]
  130. Crean, R.D.; Crane, N.A.; Mason, B.J. An evidence based review of acute and longterm effects of cannabis use on executive cognitive functions. J. Addict. Med. 2011, 5, 1. [Google Scholar] [CrossRef] [PubMed]
  131. Bossong, M.G.; Jansma, J.M.; van Hell, H.H.; Jager, G.; Oudman, E.; Saliasi, E.; Kahn, R.S.; Ramsey, N.F. Effects of δ9-tetrahydrocannabinol on human working memory function. Biol. Psychiatry 2012, 71, 693–699. [Google Scholar] [CrossRef] [PubMed]
  132. Meier, M.H.; Caspi, A.; Ambler, A.; Harrington, H.; Houts, R.; Keefe, R.S.; McDonald, K.; Ward, A.; Poulton, R.; Moffitt, T.E. Persistent cannabis users show neuropsychological decline from childhood to midlife. Proc. Natl. Acad. Sci. USA 2012, 109, E2657–E2664. [Google Scholar] [CrossRef]
  133. ElSohly, M.A.; Chandra, S.; Radwan, M.; Majumdar, C.G.; Church, J.C. A Comprehensive Review of Cannabis Potency in the United States in the Last Decade. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2021, 6, 603–606. [Google Scholar] [CrossRef] [PubMed]
  134. Patel, S.; Hill, M.N.; Cheer, J.F.; Wotjak, C.T.; Holmes, A. The endocannabinoid system as a target for novel anxiolytic drugs. Neurosci. Biobehav. Rev. 2017, 76 Pt A, 56–66. [Google Scholar] [CrossRef]
  135. Zehra, A.; Burns, J.; Liu, C.K.; Manza, P.; Wiers, C.E.; Volkow, N.D.; Wang, G.J. Cannabis Addiction and the Brain: A Review. J. Neuroimmune Pharmacol. 2018, 13, 438–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Han, B.; Compton, W.M.; Blanco, C.; Jones, C.M. Time since first cannabis use and 12-month prevalence of cannabis use disorder among youth and emerging adults in the United States. Addiction 2019, 114, 698–707. [Google Scholar] [CrossRef] [PubMed]
  137. Chan, G.C.K.; Becker, D.; Butterworth, P.; Hines, L.; Coffey, C.; Hall, W.; Patton, G. Young-adult compared to adolescent onset of regular cannabis use: A 20-year prospective cohort study of later consequences. Drug Alcohol. Rev. 2021, 40, 627–636. [Google Scholar] [CrossRef]
  138. Karila, L.; Roux, P.; Rolland, B.; Benyamina, A.; Reynaud, M.; Aubin, H.J.; Lançon, C. Acute and long-term effects of cannabis use: A review. Curr. Pharm. Des. 2014, 20, 4112–4118. [Google Scholar] [CrossRef]
  139. Katz, G.; Lobel, T.; Tetelbaum, A.; Raskin, S. Cannabis Withdrawal—A New Diagnostic Category in DSM-5. Isr. J. Psychiatry Relat. Sci. 2014, 51, 270–275. [Google Scholar]
  140. Copersino, M.L.; Boyd, S.J.; Tashkin, D.P.; Huestis, M.A.; Heishman, S.J.; Dermand, J.C.; Simmons, M.S.; Gorelick, D.A. Cannabis withdrawal among non-treatment-seeking adult cannabis users. Am. J. Addict. 2006, 15, 8–14. [Google Scholar] [CrossRef]
  141. Cornelius, J.R.; Chung, T.; Martin, C.; Wood, D.S.; Clark, D.B. Cannabis withdrawal is common among treatment-seeking adolescents with cannabis dependence and major depression, and is associated with rapid relapse to dependence. Addict. Behav. 2008, 33, 1500–1505. [Google Scholar] [CrossRef]
  142. Levin, F.R.; Brooks, D.J.; Bisaga, A.; Raby, W.; Rubin, E.; Aharonovich, E.; Nunes, E.V. Severity of dependence and motivation for treatment: Comparison of marijuana- and cocaine-dependent treatment seekers. J. Addict. Dis. 2006, 25, 33–41. [Google Scholar] [CrossRef]
  143. Bahji, A.; Stephenson, C.; Tyo, R.; Hawken, E.R.; Seitz, D.P. Prevalence of Cannabis Withdrawal Symptoms Among People With Regular or Dependent Use of Cannabinoids A Systematic Review and Meta-analysis. JAMA Netw. Open 2020, 3, e202370. [Google Scholar] [CrossRef] [PubMed]
  144. Fergusson, D.M.; Boden, J.M.; Horwood, L.J. Cannabis use and other illicit drug use: Testing the cannabis gateway hypothesis. Addiction 2006, 101, 556–569. [Google Scholar] [CrossRef] [PubMed]
  145. Madras, B.K. Are THC Levels in Oral Fluids and Blood Plasma Comparable after Oral Ingestion of Edibles Containing Cannabis or THC? Clin. Chem. 2017, 63, 629–631. [Google Scholar] [CrossRef] [PubMed]
  146. Hua, T.; Vemuri, K.; Pu, M.; Qu, L.; Han, G.W.; Wu, Y.; Zhao, S.; Shui, W.; Li, S.; Korde, A.; et al. Crystal structure of the human cannabinoid CB1. Cell 2016, 167, 750–762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Shao, Z.; Yin, J.; Chapman, K.; Grzemska, M.; Clark, L.; Wang, J.; Rosenbaum, D.M. High-resolution crystal structure of the human CB1 cannabinoid receptor. Nature 2016, 540, 602–606. [Google Scholar] [CrossRef]
  148. Katsidoni, V.; Kastellakis, A.; Panagis, G. Biphasic effects of Δ9-tetrahydrocannabinol on brain stimulation reward and motor activity. Int. J. Neuropsychopharmacol. 2013, 16, 2273–2284. [Google Scholar] [CrossRef]
  149. Vann, R.E.; Gamage, T.F.; Warner, J.A.; Marshall, E.M.; Taylor, N.L.; Martin, B.R.; Wiley, J.L. Divergent effects of cannabidiol on the discriminative stimulus and place conditioning effects of Δ9-tetrahydrocannabinol. Drug Alcohol Depend. 2008, 94, 191–198. [Google Scholar] [CrossRef]
  150. Schoeler, T.; Theobald, D.; Pingault, J.-B.; Farrington, D.P.; Coid, J.W.; Bhattacharyya, S. Developmental sensitivity to cannabis use patterns and risk for major depressive disorder in mid-life: Findings from 40 years of follow-up. Psychol. Med. 2018, 48, 2169–2176. [Google Scholar] [CrossRef]
  151. Feingold, D.; Weiser, M.; Rehm, J.; Lev-Ran, S. The association between cannabis use and mood disorders: A longitudinal study. J. Affect. Disord. 2015, 172, 211–218. [Google Scholar] [CrossRef] [PubMed]
  152. Feingold, D.; Weiser, M.; Rehm, J.; Lev-Ran, S. The association between cannabis use and anxiety disorders: Results from a population-based representative sample. Eur. Neuropsychopharmacol. 2016, 26, 493–505. [Google Scholar] [CrossRef]
  153. Danielsson, A.-K.; Lundin, A.; Agardh, E.; Allebeck, P.; Forsell, Y. Cannabis use, depression and anxiety: A 3-year prospective population-based study. J. Affect. Disord. 2016, 193, 103–108. [Google Scholar] [CrossRef]
  154. Cougle, J.R.; Hakes, J.K.; Macatee, R.J.; Chavarria, J.; Zvolensky, M.J. Quality of life and risk of psychiatric disorders among regular users of alcohol, nicotine, and cannabis: An analysis of the national epidemiological survey on alcohol and related conditions (nesarc). J. Psychiatr. Res. 2015, 66, 135–141. [Google Scholar] [CrossRef] [PubMed]
  155. Østergaard, M.L.; Nordentoft, M.; Hjorthøj, C. Associations between substance use disorders and suicide or suicide attempts in people with mental illness: A danish nation-wide, prospective, register-based study of patients diagnosed with schizophrenia, bipolar disorder, unipolar depression or personality disorder. Addiction 2017, 112, 1250–1259. [Google Scholar]
  156. Van Dam, N.T.; Bedi, G.; Earleywine, M. Characteristics of clinically anxious versus non-anxious regular, heavy marijuana users. Addict. Behav. 2012, 37, 1217–1223. [Google Scholar] [CrossRef]
  157. Sagar, K.A.; Dahlgren, M.K.; Racine, M.T.; Dreman, M.W.; Olson, D.P.; Gruber, S.A. Joint effects: A pilot investigation of the impact of bipolar disorder and marijuana use on cognitive function and mood. PLoS ONE 2016, 11, e0157060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Hjorthøj, C.; Østergaard, M.L.D.; Benros, M.E.; Toftdahl, N.G.; Erlangsen, A.; Andersen, J.T.; Nordentoft, M. Association between alcohol and substance use disorders and allcause and cause-specific mortality in schizophrenia, bipolar disorder, and unipolar depression: A nationwide, prospective, register-based study. Lancet Psychiatry 2015, 2, 801–808. [Google Scholar] [CrossRef]
  159. Duperrouzel, J.; Hawes, S.W.; Lopez-Quintero, C.; Pacheco-Colón, I.; Comer, J.; Gonzalez, R. The association between adolescent cannabis use and anxiety: A parallel process analysis. Addict. Behav. 2018, 78, 107–113. [Google Scholar] [CrossRef] [PubMed]
  160. Copeland, J.; Rooke, S.; Swift, W. Changes in cannabis use among young people: Impact on mental health. Curr. Opin. Psychiatry 2013, 26, 325–329. [Google Scholar] [CrossRef]
  161. Degenhardt, L.; Coffey, C.; Romaniuk, H.; Swift, W.; Carlin, J.B.; Hall, W.D.; Patton, G.C. The persistence of the association between adolescent cannabis use and common mental disorders into young adulthood. Addiction 2013, 108, 124–133. [Google Scholar] [CrossRef] [PubMed]
  162. Hayatbakhsh, M.R.; Najman, J.M.; Jamrozik, K.; Mamun, A.A.; Alati, R.; Bor, W. Cannabis and anxiety and depression in young adults: A large prospective study. J. Am. Acad. Child Adolesc. Psychiatry 2007, 46, 408–417. [Google Scholar] [CrossRef]
  163. Earleywine, M.; Bolles, J.R. Marijuana, expectancies, and post-traumatic stress symptoms: A preliminary investigation. J. Psychoact. Drugs 2014, 46, 171–177. [Google Scholar] [CrossRef] [PubMed]
  164. Manhapra, A.; Stefanovics, E.; Rosenheck, R. Treatment outcomes for veterans with PTSD and substance use: Impact of specific substances and achievement of abstinence. Drug Alcohol Depend. 2015, 156, 70–77. [Google Scholar] [CrossRef] [PubMed]
  165. National Academies of Sciences, Engineering, and Medicine. The Health Effects of Cannabis and Cannabinoids: The Current State of Evidence and Recommendations for Research; The National Academies Press: Washington, DC, USA, 2017. [Google Scholar]
  166. Campeny, E.; López-Pelayo, H.; Nutt, D.; Blithikioti, C.; Oliveras, C.; Nuño, L.; Maldonado, R.; Florez, G.; Arias, F.; Fernández-Artamendi, S.; et al. The blind men and the elephant: Systematic review of systematic reviews of cannabis use related health harms. Eur. Neuropsychopharmacol. 2020, 33, 1–35. [Google Scholar] [CrossRef]
  167. Ney, L.J.; Matthews, A.; Bruno, R.; Felmingham, K.L. Cannabinoid interventions for PTSD: Where to next? Prog. Neuropsychopharmacol. Biol. Psychiatry 2019, 93, 124–140. [Google Scholar] [CrossRef] [PubMed]
  168. Rock, E.M.; Parker, L.A. Constituents of Cannabis Sativa. In Cannabinoids and Neuropsychiatric Disorders. Advances in Experimental Medicine and Biology; Murillo-Rodriguez, E., Pandi-Perumal, S.R., Monti, J.M., Eds.; Springer: Cham, Switzerland, 2021; Volume 1264. [Google Scholar]
  169. Ahmed, M.; Boileau, I.; Le Foll, B.; Carvalho, A.F.; Kloiber, S. The endocannabinoid system in social anxiety disorder: From pathophysiology to novel therapeutics. Braz. J. Psychiatry 2022, 44, 81–93. [Google Scholar] [CrossRef] [PubMed]
  170. Dos Santos, R.G.; Hallak, J.E.; Crippa, J.A.S. Neuropharmacological effects of the main phytocannabinoids: A narrative review. In Cannabinoids and Neuropsychiatric Disorders; Springer: Berlin/Heidelberg, Germany, 2021; pp. 29–45. [Google Scholar]
  171. Stanciu, C.N.; Brunette, M.F.; Teja, N.; Budney, A.J. Evidence for Use of Cannabinoids in Mood Disorders, Anxiety Disorders, and PTSD: A Systematic Review. Psychiatr. Serv. 2021, 72, 429–436. [Google Scholar] [CrossRef]
  172. Orsolini, L.; Chiappini, S.; Volpe, U.; Berardis, D.; Latini, R.; Papanti, G.D.; Corkery, A.J.M. Use of Medicinal Cannabis and Synthetic Cannabinoids in Post-Traumatic Stress Disorder (PTSD): A Systematic Review. Medicina 2019, 55, 525. [Google Scholar] [CrossRef]
  173. Bhattacharyya, S.; Morrison, P.D.; Fusar-Poli, P.; Martin-Santos, R.; Borgwardt, S.; Winton-Brown, T.; Nosarti, C.; O’Carroll, C.M.; Seal, M.; Allen, P. Opposite effects of delta-9-tetrahydrocannabinol and cannabidiol on human brain function and psychopathology. Neuropsychopharmacology 2010, 35, 764–774. [Google Scholar] [CrossRef]
  174. Bossong, M.G.; Jansma, J.M.; van Hell, H.H.; Jager, G.; Kahn, R.S.; Ramsey, N.F. Default mode network in the effects of Δ9-Tetrahydrocannabinol (THC) on human executive function. PLoS ONE 2013, 8, e70074. [Google Scholar] [CrossRef] [PubMed]
  175. Fusar-Poli, P.; Allen, P.; Bhattacharyya, S.; Crippa, J.A.; Mechelli, A.; Borgwardt, S.; Martin-Santos, R.; Seal, M.L.; O’Carrol, C.; Atakan, Z.; et al. Modulation of effective connectivity during emotional processing by Δ9-tetrahydrocannabinol and cannabidiol. Int. J. Neuropsychopharmacol. 2010, 13, 421–432. [Google Scholar] [CrossRef] [PubMed]
  176. Karniol, I.G.; Shirakawa, I.; Kasinski, N.; Pfeferman, A.; Carlini, E.A. Cannabidiol interferes with the effects of delta 9-tetrahydrocannabinol in man. Eur. J. Pharmacol. 1974, 28, 172–177. [Google Scholar] [CrossRef]
  177. Martin-Santos, R.; a Crippa, J.; Batalla, A.; Bhattacharyya, S.; Atakan, Z.; Borgwardt, S.; Allen, P.; Seal, M.; Langohr, K.; McGuire, K.P.; et al. Acute effects of a single, oral dose of d9-tetrahydrocannabinol (THC) and cannabidiol (CBD) administration in healthy volunteers. Curr. Pharm. Des. 2012, 18, 4966–4979. [Google Scholar] [CrossRef]
  178. Turna, J.; Patterson, B.; Van Ameringen, M. Is cannabis treatment for anxiety, mood, and related disorders ready for prime time? Depress. Anxiety 2017, 34, 1006–1017. [Google Scholar] [CrossRef]
  179. Gallego-Landin, I.; García-Baos, A.; Castro-Zavala, A.; Valverde, O. Reviewing the Role of the Endocannabinoid System in the Pathophysiology of Depression. Front. Pharmacol. 2021, 12, 762738. [Google Scholar] [CrossRef]
  180. Nelson, K.; Walsh, D.; Deeter, P.; Sheehan, F. A phase II study of delta-9-tetrahydrocannabinol for appetite stimulation in cancer-associated anorexia. J. Palliat Care 1994, 10, 14–18. [Google Scholar]
  181. Chang, A.E.; Shiling, D.J.; Stillman, R.C.; Goldberg, N.H.; Seipp, C.A.; Barofsky, I.; Simon, R.M.; Rosenberg, S.A. Delta-9-tetrahydrocannabinol as an antiemetic in cancer patients receiving high-dose methotrexate. A prospective, randomized evaluation. Ann. Intern. Med. 1979, 91, 819–824. [Google Scholar] [CrossRef]
  182. Frytak, S.; Moertel, C.G.; O’Fallon, J.R.; Rubin, J.; Creagan, E.T.; O’Connell, M.J.; Schutt, A.J.; Schwartau, N.W. Delta-9-tetrahydrocannabinol as an antiemetic for patients receiving cancer chemotherapy. A comparison with prochlorperazine and a placebo. Ann. Intern. Med. 1979, 91, 825–830. [Google Scholar] [CrossRef]
  183. Costa, B.; Comelli, F. Pain. In Handbook of Cannabis; Pertwee, R.G., Ed.; Oxford University Press: Oxford, UK, 2014; pp. 473–486. [Google Scholar]
  184. Ware, M.A.; Wang, T.; Shapiro, S.; Robinson, A.; Ducruet, T.; Huynh, T.; Gamsa, A.; Bennett, G.J.; Collet, J.-P. Smoked cannabis for chronic neuropathic pain: A randomized controlled trial. CMAJ 2010, 182, E694–E701. [Google Scholar] [CrossRef]
  185. Wilsey, B.; Marcotte, T.; Deutsch, R.; Gouaux, B.; Sakai, S.; Donaghe, H. Low-dose vaporized cannabis significantly improves neuropathic pain. J. Pain 2013, 14, 136–148. [Google Scholar] [CrossRef]
  186. Wilsey, B.; Marcotte, T.D.; Deutsch, R.; Zhao, H.; Prasad, H.; Phan, A. An Exploratory Human Laboratory Experiment Evaluating Vaporized Cannabis in the Treatment of Neuropathic Pain From Spinal Cord Injury and Disease. J. Pain 2016, 17, 982–1000. [Google Scholar] [CrossRef]
  187. Betthauser, K.; Pilz, J.; Vollmer, L.E. Use and effects of cannabinoids in military veterans with posttraumatic stress disorder. Am. J. Health Syst. Pharm. 2015, 72, 1279–1284. [Google Scholar] [CrossRef]
  188. Jetly, R.; Heber, A.; Fraser, G.; Boisvert, D. The efficacy of nabilone, a synthetic cannabinoid, in the treatment of PTSD-associated nightmares: A preliminary randomized, double-blind, placebo-controlled cross-over design study. Psychoneuroendocrinology 2015, 51, 585–588. [Google Scholar] [CrossRef] [PubMed]
  189. Rock, E.M.; Parker, L.A. Cannabinoids As Potential Treatment for Chemotherapy-Induced Nausea and Vomiting. Front. Pharmacol. 2016, 7, 221. [Google Scholar] [CrossRef] [PubMed]
  190. Herman, T.S.; Jones, S.E.; Dean, J.; Leigh, S.; Dorr, R.; E Moon, T.; Salmon, E.S. Nabilone: A potent antiemetic cannabinol with minimal euphoria. Biomedicine 1977, 27, 331–334. [Google Scholar] [PubMed]
  191. Beal, J.E.; Olson, R.; Laubenstein, L.; Morales, J.O.; Bellman, P.; Yangco, B.; Lefkowitz, L.; Plasse, T.F.; Shepard, K.V.; John, S. Dronabinol as a Treatment for Anorexia Associated with Weight Loss in Patients with AIDS. J. Pain Symptom Manag. 1995, 10, 89–97. [Google Scholar] [CrossRef]
  192. Badowski, M.E.; Yanful, P.K. Dronabinol oral solution in the management of anorexia and weight loss in AIDS and cancer. Ther. Clin. Risk Manag. 2018, 14, 643–651. [Google Scholar] [CrossRef] [Green Version]
  193. Parker, L.A.; Mechoulam, R.; Schlievert, C.; Abbott, L.; Fudge, M.L.; Burton, P. Effects of cannabinoids on lithium-induced conditioned rejection reactions in a rat model of nausea. Psychopharmacology 2003, 166, 156–162. [Google Scholar] [CrossRef]
  194. Cluny, N.L.; Naylor, R.J.; Whittle, B.A.; Javid, F.A. The effects of cannabidiol and tetrahydrocannabinol on motion-induced emesis in Suncus murinus. Basic Clin. Pharmacol. Toxicol. 2008, 103, 150–156. [Google Scholar] [CrossRef]
  195. Parker, L.A.; Kwiatkowska, M.; Burton, P.; Mechoulam, R. Effect of cannabinoids on lithium-induced vomiting in the Suncus murinus (house musk shrew). Psychopharmacology 2004, 171, 156–161. [Google Scholar] [CrossRef]
  196. van Vliet, S.A.; Vanwersch, R.A.; Jongsma, M.J.; Olivier, B.; Philippens, I.H. Therapeutic effects of delta9-THC and modafinil in a marmoset Parkinson model. Eur. Neuropsychopharmacol. 2008, 18, 383–389. [Google Scholar] [CrossRef]
  197. Raman, C.; McAllister, S.D.; Rizvi, G. Amyotrophic lateral sclerosis: Delayed disease progression in mice by treatment with a cannabinoid. Amyotroph. Lateral Scler. Other Motor Neuron Disord. 2004, 5, 33–39. [Google Scholar] [CrossRef] [PubMed]
  198. Crippa, J.A.; Guimarães, F.S.; Campos, A.C.; Zuardi, A.W. Translational investigation of the therapeutic potential of cannabidiol (CBD): Toward a new age. Front. Immunol. 2018, 9, 2009. [Google Scholar] [CrossRef] [PubMed]
  199. Rock, E.M.; Bolognini, D.; Limebeer, C.L.; Cascio, M.G.; Anavi-Goffer, S.; Fletcher, P.J.; Mechoulam, R.; Pertwee, R.G.; Parker, L.A. Cannabidiol, a non-psychotropic component of cannabis, attenuates vomiting and nausea-like behaviour via indirect agonism of 5-HT(1A) somatodendritic autoreceptors in the dorsal raphe nucleus. Br. J. Pharmacol. 2012, 165, 2620–2634. [Google Scholar] [CrossRef]
  200. Fraguas-Sánchez, A.I.; Torres-Suárez, A.I. Medical use of cannabinoids. Drugs 2018, 78, 1665–1703. [Google Scholar] [CrossRef]
  201. Jacobs, D.S.; Kohut, S.J.; Jiang, S.; Nikas, S.P.; Makriyannis, A.; Bergman, J. Acute and chronic effects of cannabidiol on Δ9-tetrahydrocannabinol (Δ9-THC)-induced disruption in stop signal task performance. Exp. Clin. Psychopharmacol. 2016, 24, 320–330. [Google Scholar] [CrossRef]
  202. Zuardi, A.W.; Cosme, R.A.; Graeff, F.G.; Guimaraes, F.S. Effects of ipsapirone and cannabidiol on human experimental anxiety. J. Psychopharmacol. 1993, 7 (Suppl. S1), 82–88. [Google Scholar] [CrossRef]
  203. Linares, I.M.; Zuardi, A.W.; Pereira, L.C.; Queiroz, R.H.; Mechoulam, R.; Guimarães, F.S.; Crippa, J.A. Cannabidiol presents an inverted U-shaped dose-response curve in a simulated public speaking test. Braz. J. Psychiatry 2019, 41, 9–14. [Google Scholar] [CrossRef]
  204. Nader, D.A.; Sanchez, Z.M. Effects of regular cannabis use on neurocognition, brain structure, and function: A systematic review of findings in adults. Am. J. Drug Alcohol Abuse 2018, 44, 4–18. [Google Scholar] [CrossRef] [PubMed]
  205. Chagas, M.H.; Zuardi, A.W.; Tumas, V.; Pena-Pereira, M.A.; Sobreira, E.T.; Bergamaschi, M.M.; Dos Santos, A.C.; Teixeira, A.L.; Hallak, J.E.C.; Crippa, J.A.S. Effects of cannabidiol in the treatment of patients with Parkinson’s disease: An exploratory double-blind trial. J. Psychopharmacol. 2014, 28, 1088–1098. [Google Scholar] [CrossRef]
  206. Bergamaschi, M.M.; Queiroz, R.H.; Chagas, M.H.; De Oliveira, D.C.G.; De Martinis, B.; Kapczinski, F.; de Quevedo, J.L.; Roesler, R.; Schroder, N.; Nardi, A.E.; et al. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naive social phobia patients. Neuropsychopharmacology 2011, 36, 1219–1226. [Google Scholar] [CrossRef]
  207. Crippa, J.A.; Derenusson, G.N.; Ferrari, T.B.; Wichert-Ana, L.; Duran, F.L.; Martin-Santos, R.; Simões, M.V.; Bhattacharyya, S.; Fusar-Poli, P.; Atakan, Z.; et al. Neural basis of anxiolytic effects of cannabidiol (CBD) in generalized social anxiety disorder: A preliminary report. J. Psychopharmacol. 2011, 25, 121–130. [Google Scholar] [CrossRef] [PubMed]
  208. Rock, E.M.; Limebeer, C.L.; Parker, L.A. Effect of combined doses of Δ(9)-tetrahydrocannabinol (THC) and cannabidiolic acid (CBDA) on acute and anticipatory nausea using rat (Sprague- Dawley) models of conditioned gaping. Psychopharmacology 2015, 232, 4445–4454. [Google Scholar] [CrossRef] [PubMed]
  209. Rock, E.M.; Limebeer, C.L.; Parker, L.A. Effect of cannabidiolic acid and Δ9-tetrahydrocannabinol on carrageenan-induced hyperalgesia and edema in a rodent model of inflammatory pain. Psychopharmacology 2018, 235, 3259–3271. [Google Scholar] [CrossRef] [PubMed]
  210. Rock, E.M.; Limebeer, C.L.; Petrie, G.N.; Williams, L.A.; Mechoulam, R.; Parker, L.A. Effect of prior foot shock stress and Δ9-tetrahydrocannabinol, cannabidiolic acid, and cannabidiol on anxiety-like responding in the light-dark emergence test in rats. Psychopharmacology 2017, 234, 2207–2217. [Google Scholar] [CrossRef] [PubMed]
  211. Skrabek, R.Q.; Galimova, L.; Ethans, K.; Perry, D. Nabilone for the treatment of pain in fibromyalgia. J. Pain 2008, 9, 164–173. [Google Scholar] [CrossRef]
  212. Maida, V.; Ennis, M.; Irani, S.; Corbo, M.; Dolzhykov, M. Adjunctive nabilone in cancer pain and symptom management: A prospective observational study using propensity scoring. J. Support Oncol. 2008, 6, 119–124. [Google Scholar]
  213. Weber, J.; Schley, M.; Casutt, M.; Gerber, H.; Schuepfer, G.; Rukwied, R.; Schleinzer, W.; Ueberall, M.; Konrad, C. Tetrahydrocannabinol (Delta 9-THC) treatment in chronic central neuropathic pain and fibromyalgia patients: Results of a multicenter survey. Anesthesiol. Res. Pract. 2009, 2009, 827290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Schley, M.; Legler, A.; Skopp, G.; Schmelz, M.; Konrad, C.; Rukwied, R. Delta-9-THC based monotherapy in fibromyalgia patients on experimentally induced pain, axon reflex flare, and pain relief. Curr. Med. Res. Opin. 2006, 22, 1269–1276. [Google Scholar] [CrossRef]
  215. Phan, K.L.; Angstadt, M.; Golden, J.; Onyewuenyi, I.; Popovska, A.; de Wit, H. Cannabinoid modulation of amygdala reactivity to social signals of threat in humans. J. Neurosci. 2008, 28, 2313–2319. [Google Scholar] [CrossRef]
  216. Colizzi, M.; Ruggeri, M.; Bhattacharyya, S. Unraveling the Intoxicating and Therapeutic Effects of Cannabis Ingredients on Psychosis and Cognition. Front. Psychol. 2020, 11, 833. [Google Scholar] [CrossRef]
  217. Rabinak, C.A.; Angstadt, M.; Sripada, C.S.; Abelson, J.L.; Liberzon, I.; Milad, M.R.; Phan, K.L. Cannabinoid facilitation of fear extinction memory recall in humans. Neuropharmacology 2013, 64, 396–402. [Google Scholar] [CrossRef]
  218. Rabinak, C.A.; Phan, K.L. Cannabinoid modulation of fear extinction brain circuits: A novel target to advance anxiety treatment. Curr. Pharm. 2014, 20, 2212–2217. [Google Scholar] [CrossRef] [PubMed]
  219. Fabre, L.F.; Mclendon, D.A.V.I.D. The efficacy and safety of nabilone (a synthetic cannabinoid) in the treatment of anxiety. J. Clin. Pharmacol. 1981, 21, 377S–382S. [Google Scholar] [CrossRef] [PubMed]
  220. Lee, J.Y.; Brook, J.S.; Finch, S.J.; Brook, D.W. Trajectories of cannabis use beginning in adolescence associated with symptoms of posttraumatic stress disorder in the midthirties. Subst. Abuse 2018, 39, 39–45. [Google Scholar] [CrossRef] [PubMed]
  221. Withey, S.L.; Kangas, B.D.; Charles, S.; Gumbert, A.B.; Eisold, J.E.; George, S.R.; Bergman, J.; Madras, B.K. Effects of daily Δ9-Tetrahydrocannabinol (THC) alone or combined with cannabidiol (CBD) on cognition-based behavior and activity in adolescent nonhuman primates. Drug Alcohol Depend. 2021, 221, 108629. [Google Scholar] [CrossRef]
  222. Hasbi, A.; Madras, B.K.; Bergman, J.; Kohut, S.; Lin, Z.; Withey, S.L.; George, S.R. Δ-9-Tetrahydrocannabinol Increases Dopamine D1-D2 Receptor Heteromer and Elicits Phenotypic Reprogramming in Adult Primate Striatal Neurons. IScience 2020, 23, 100794. [Google Scholar] [CrossRef]
  223. Withey, S.L.; Bergman, J.; Huestis, M.A.; George, S.R.; Madras, B.K. THC and CBD blood and brain concentrations following daily administration to adolescent primates. Drug Alcohol Depend. 2020, 213, 108129. [Google Scholar] [CrossRef]
  224. Englund, A.; Freeman, T.P.; Murray, R.M.; McGuire, P. Can we make cannabis safer? Lancet Psychiatry 2017, 4, 643–648. [Google Scholar] [CrossRef]
  225. Schubart, C.D.; Sommer, I.E.; van Gastel, W.A.; Goetgebuer, R.L.; Kahn, R.S.; Boks, M.P. Cannabis with high cannabidiol content is associated with fewer psychotic experiences. Schizophr. Res. 2011, 130, 216–221. [Google Scholar] [CrossRef]
  226. Colizzi, M.; Bhattacharyya, S. Does cannabis composition matter? Differential effects of delta-9-tetrahydrocannabinol and cannabidiol on human cognition. Curr. Addict. Rep. 2017, 4, 62–74. [Google Scholar] [CrossRef]
  227. Sarne, Y. Beneficial and deleterious effects of cannabinoids in the brain: The case of ultra-low dose of THC. Am. J. Drugs Alcohol Abuse 2019, 45, 551–562. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the endocannabinoid signaling system. Endocannabinoids (eCBs) 2AG (2-arachidonoylglycerol) and AEA (anandamide) are synthesized in postsynaptic neurons and act pre-synaptically at their receptors (CB1Rs) in a retrograde mode (A). eCBs can also activate CB1R localized post-synaptically in some cases. In general, activation of CB1 receptors by eCBs decreases the probability of neurotransmitter release through multiple mechanisms, comprising inhibition of calcium influx and activation of potassium channels These effects are terminated by eCB reuptake followed by degradation: 2AG by MAGL pre-synaptically and AEA by FAAH post-synaptically. A large proportion (45–48%) of synapses in different brain regions is tripartite (B), formed by pre-synaptic and post-synaptic neurons, and a glial cell—an astrocyte—which expresses CB1R. In response to neuronal activation, eCBs released by the postsynaptic neurons can activate CB1R expressed on astrocytes. The astrocytes respond with an increase in intracellular calcium leading to release of gliotransmitters (B).
Figure 1. Schematic representation of the endocannabinoid signaling system. Endocannabinoids (eCBs) 2AG (2-arachidonoylglycerol) and AEA (anandamide) are synthesized in postsynaptic neurons and act pre-synaptically at their receptors (CB1Rs) in a retrograde mode (A). eCBs can also activate CB1R localized post-synaptically in some cases. In general, activation of CB1 receptors by eCBs decreases the probability of neurotransmitter release through multiple mechanisms, comprising inhibition of calcium influx and activation of potassium channels These effects are terminated by eCB reuptake followed by degradation: 2AG by MAGL pre-synaptically and AEA by FAAH post-synaptically. A large proportion (45–48%) of synapses in different brain regions is tripartite (B), formed by pre-synaptic and post-synaptic neurons, and a glial cell—an astrocyte—which expresses CB1R. In response to neuronal activation, eCBs released by the postsynaptic neurons can activate CB1R expressed on astrocytes. The astrocytes respond with an increase in intracellular calcium leading to release of gliotransmitters (B).
Brainsci 13 00325 g001
Figure 2. Modulation of neuronal activity by the endocannabinoid system. Endocannabinoids modulate neuronal activity as illustrated for dopamine (DA) neurons with cell bodies in the ventral tegmental area (VTA). CB1 receptors on GABAergic terminals (A) can facilitate dopaminergic activity through suppression of the inhibitory input onto the GABA receptors present on DA neurons, leading to an increase of DA release. Further, the activation of mGluR5 receptor by released glutamate (B) leads to the synthesis and release of 2-arachidonoylglycerol (2-AG), which then retrogradely activates CB1 receptors to inhibit additional glutamate release.
Figure 2. Modulation of neuronal activity by the endocannabinoid system. Endocannabinoids modulate neuronal activity as illustrated for dopamine (DA) neurons with cell bodies in the ventral tegmental area (VTA). CB1 receptors on GABAergic terminals (A) can facilitate dopaminergic activity through suppression of the inhibitory input onto the GABA receptors present on DA neurons, leading to an increase of DA release. Further, the activation of mGluR5 receptor by released glutamate (B) leads to the synthesis and release of 2-arachidonoylglycerol (2-AG), which then retrogradely activates CB1 receptors to inhibit additional glutamate release.
Brainsci 13 00325 g002
Table 1. Examples of preclinical studies showing the effects of cannabinoids on depression-like and anxiety-like behaviors.
Table 1. Examples of preclinical studies showing the effects of cannabinoids on depression-like and anxiety-like behaviors.
SpeciesContext and Cannabinoid UsedSummary of ResultsReferences
Rodents
(rat, mice)
CB1 antagonist SR141716
Depression- or anxiety-like behavior
Depressive-like phenotype[80,81]
Rodents
(rat, mice)
CB1 receptor antagonists, Rimonabant and AM251anxiogenic effects[80,81,82]
Rodents
(rat, mice)
Rimonabant
Depression- or anxiety-like behavior
No effect[80,83,84,85]
Rodents
(rat, mice)
Rimonabant
Depression- or anxiety-like behavior
Anxiolytic effect[80]
Rodents
(rat, mice)
CB1 agonists HU210 or oleamide
Depression- or anxiety-like behavior
Anxiolytic and/or antidepression-like behavior[10,68]
Rodents
(rat, mice)
CB1 agonist THC
Depression- or anxiety-like behavior
Anxiolytic and/or antidepression-like behavior[10,69,70,80]
Rodents
(rat, mice)
Synthetic CB1 agonists
WIN55,212-2; CP55,940
Depression- or anxiety-like behavior
Anxiolytic and/or antidepression-like behavior[10,71,72,80]
Rodents
(rat, mice)
Higher doses of CB1 agonists
Depression- or anxiety-like behavior
Pro-depressive and anxiogenic[69,75,76,77,78,80]
Rodents
(rat, mice)
The endogenous AEA
Depression- or anxiety-like behavior
Anxiolytic and/or antidepression-like behavior[73,74,80]
C57BL/6 miceCBD
Chronic stress
Treatment precludes anxiety-like behaviorreviewed in [80]
C57BL/6 miceWIN55,212-2
Stress-induced anxiety
Reversal of social stress-induced anxiety-like phenotypereviewed in [80]
C57BL/6 miceMAGL inhibitor
Chronic stress
Enhanced adult neurogenesis involved in anxiolytic like effectsreviewed in [80]
CB1 KO miceRole of CB1 receptor in social anxiety and memoryAcute CB1 antagonist AM251 increased anxiety-like behavior in CB1 KOs and WT reviewed in [80]
CB1 KO miceRole of CB1 receptor in anxietyAnxiogenic phenotype only under high light conditionsreviewed in [80]
CB1 KO miceRole of CB1 receptor in stress coping behaviorsIncreased passive stress coping behaviorreviewed in [80]
CB1 KO miceBehavioral characterization of CB1-KO miceIncreased anxiety-like behavior. Reversed by cannabinoid antagonistreviewed in [80]
DAGL-α KO miceRole of DAGL-α, and 2-AG in anxiety-like behaviorEnhanced anxiety, stress and fear responses in DAGL-α KOreviewed in [80]
See [10,67,68,69,70,71,72,73,74,75,76,77,78,80,81,82,83,84,85] for more details.
Table 2. Examples of cannabinoid-based therapies to treat anxiety and/or depression.
Table 2. Examples of cannabinoid-based therapies to treat anxiety and/or depression.
Cannabinoid-Based DrugDosage
(Where Indicated)
Outcomes for Anxiety/DepressionRelated ConditionSystematic Reviews
(References)
CannabisNA↓AnxietyHIVHuang et al., 2016
Ref. [5]
CannabisNA↓Anxiety
↓Depression
↑Quality of life
Antidepressant, anxiolytic and quality of lifeBlack et al., 2019
Chadwick et al., 2020
Refs. [108,109]
Cannabis sativaNA↓Anxiety
↓Depression
AntidepressantBlack et al., 2019
Ref. [109]
NabiloneNA↓AnxietyFibromyalgiaHuang et al., 2016
Ref. [5]
NabiloneNA↓Anxiety and StressCancerHuang et al., 2016
Ref. [5]
Nabilone28 days of 1 mg↓AnxietyPsychoneurotic anxiety disorderStanciu et al., 2021
Ref. [171]
Nabiloneonce 2 mg, then weekly 0.5–5 mg for five weeksNo improvement in anxiety.Anxiety neurosis or generalized anxiety disorderStanciu et al., 2021
Ref. [171]
Dronabinol [THC]NA↓AnxietyChronic neuropathic painHuang et al., 2016
Ref. [5]
Dronabinol (oral)16.6 mg (7.5–25 mg)↓Anxiety
↓Depression
↑Quality of life
Analgesic and impact on mental health outcomesBlack et al., 2019
Ref. [109]
Dronabinol (oral)5–10 mg↓Depression
↑Quality of life
Antidepressant and quality of lifeBlack et al., 2019
Chadwick et al., 2020
Refs. [108,109]
Dronabinol (oral)10 mg (2.5–10 mg)↓Anxiety
↑Quality of life
Antidepressant and quality of lifeBlack et al., 2019
Chadwick et al., 2020
Refs. [108,109]
THC7 days of 0.3 mg/kgNo improvement in depressive symptoms.Unipolar and bipolar major depressionStanciu et al., 2021
Ref. [171]
CBD4 weeks of 300 mg↓AnxietySocial anxiety disorderStanciu et al., 2021
Ref. [171]
CBDSingle dose of 600 mg↓AnxietySocial anxiety disorderStanciu et al., 2021
Ref. [171]
CBDSingle dose of 400 mg↓AnxietySocial anxiety disorderStanciu et al., 2021
Ref. [171]
Sativex [THC+CBD]NA↓Anxiety
↑Quality of life
Diabetic-neuropathyHuang et al., 2016
Ref. [5]
CBD + THCNASome but not all studies:
↓Anxiety
↓Depression
↓Stress
↑Quality of life
PTSDOrsolini et al., 2019
Ref. [172]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hasbi, A.; Madras, B.K.; George, S.R. Endocannabinoid System and Exogenous Cannabinoids in Depression and Anxiety: A Review. Brain Sci. 2023, 13, 325. https://doi.org/10.3390/brainsci13020325

AMA Style

Hasbi A, Madras BK, George SR. Endocannabinoid System and Exogenous Cannabinoids in Depression and Anxiety: A Review. Brain Sciences. 2023; 13(2):325. https://doi.org/10.3390/brainsci13020325

Chicago/Turabian Style

Hasbi, Ahmed, Bertha K. Madras, and Susan R. George. 2023. "Endocannabinoid System and Exogenous Cannabinoids in Depression and Anxiety: A Review" Brain Sciences 13, no. 2: 325. https://doi.org/10.3390/brainsci13020325

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop