Next Article in Journal
Prolonged Activation of Brain CB2 Signaling Modulates Hypothalamic Microgliosis and Astrogliosis in High Fat Diet-Fed Mice
Next Article in Special Issue
Cannabidiol Prevents Spontaneous Fear Recovery after Extinction and Ameliorates Stress-Induced Extinction Resistance
Previous Article in Journal
MicroRNA-29a Manifests Multifaceted Features to Intensify Radiosensitivity, Escalate Apoptosis, and Revoke Cell Migration for Palliating Radioresistance-Enhanced Cervical Cancer Progression
Previous Article in Special Issue
Pro-Inflammatory Cytokines: Potential Links between the Endocannabinoid System and the Kynurenine Pathway in Depression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modulation of Endocannabinoid System Components in Depression: Pre-Clinical and Clinical Evidence

1
Department of Psychology, School of Psychological Sciences, University of Haifa, Haifa 3498838, Israel
2
The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(10), 5526; https://doi.org/10.3390/ijms23105526
Submission received: 30 March 2022 / Revised: 9 May 2022 / Accepted: 9 May 2022 / Published: 15 May 2022

Abstract

:
Depression is characterized by continuous low mood and loss of interest or pleasure in enjoyable activities. First-line medications for mood disorders mostly target the monoaminergic system; however, many patients do not find relief with these medications, and those who do suffer from negative side effects and a discouragingly low rate of remission. Studies suggest that the endocannabinoid system (ECS) may be involved in the etiology of depression and that targeting the ECS has the potential to alleviate depression. ECS components (such as receptors, endocannabinoid ligands, and degrading enzymes) are key neuromodulators in motivation and cognition as well as in the regulation of stress and emotions. Studies in depressed patients and in animal models for depression have reported deficits in ECS components, which is motivating researchers to identify potential diagnostic and therapeutic biomarkers within the ECS. By understanding the effects of cannabinoids on ECS components in depression, we enhance our understanding of which brain targets they hit, what biological processes they alter, and eventually how to use this information to design better therapeutic options. In this article, we discuss the literature on the effects of cannabinoids on ECS components of specific depression-like behaviors and phenotypes in rodents and then describe the findings in depressed patients. A better understanding of the effects of cannabinoids on ECS components in depression may direct future research efforts to enhance diagnosis and treatment.

1. Introduction

Depression is one of the world’s most common psychiatric disorders, with a prevalence rate of 3.8%, according to the World Health Organization [1]. The lifetime prevalence of depression is as high as 20%, with a female-to-male ratio of about 5:2 [2]. Major depressive disorder (MDD) has been one of the leading causes of years lived with disability during the last three decades and it is also a major contributor to suicide deaths [1,3].
The Diagnostic and Statistical Manual of Mental Disorders (DSM-5), the guide used by health care professionals, states that the common feature of depressive disorders is the presence of sad, empty or irritable mood, accompanied by somatic and cognitive changes that significantly affect the individual’s capacity to function [4].
Depression is a complex phenomenon with many subtypes and many likely etiologies. There are multiple treatments with varying success rates, but the efficacy of currently used drugs is limited, particularly for preventing relapse and recurrence [5]. Selective serotonin reuptake inhibitors (SSRIs) and cognitive-behavioral therapy (CBT) are the two first-line treatments for depression [6]. SSRIs are among the most commonly prescribed drugs worldwide and are better tolerated than their predecessors, the tricyclic antidepressant family (TCAs); however, they have adverse side effects and when discontinued by the patient might cause withdrawal and rebound phenomena [7,8].
Many patients do not respond to SSRIs, or show intolerance to the drugs’ undesired effects [9]. About 60% of MDD patients continue to report residual impairments even after treatment with SSRIs [10], and around 33% of MDD patients develop resistance to antidepressant drugs [11]. Moreover, about 38% of patients suffer from at least one side effect, the most common of which include impaired sexual functioning, sleeping problems, and weight gain [12]. These negative side effects are common across antidepressant drug classes, and can withhold initiation of drug treatment and contribute to discontinuation of treatment [13,14]. Therefore, extensive efforts have been made to develop new approaches that treat depression with reduced side effects.
This partial success in treating depression is associated with our insufficient understanding of the underlying mechanisms of the disorder. In this sense, there is evidence to suggest that the endocannabinoid system (ECS) is impaired in MDD, providing a unique opportunity to identify potential diagnostic and therapeutic biomarkers. The ECS is a widespread neuromodulatory system involving a combination of endocannabinoids, enzymes, and cannabinoid receptors that help regulate numerous functions, including emotions and cognition.
A growing body of evidence suggests that the etiology of MDD may involve the ECS [15]. Specifically, it has been proposed that ECS deficits might have a depressive and anxiogenic effect on behavior, while elevation of ECS signaling can have antidepressant and anxiolytic properties [16,17]. Hence, some cannabis sativa plant compounds, which target the ECS, have been attracting great interest for their potential therapeutic use [18]. Recent measurements of public opinion suggest that people believe cannabis provides relief from depression and do not perceive it as harmful [19]. Due to the increase in the number of people that self-medicate with cannabis to relieve depressive symptoms, it is essential to determine whether cannabis is effective for managing depression.
Longitudinal studies have reported mixed evidence regarding the association between cannabis use and depression [20,21]. Some suggest that cannabis use may increase the risk for developing depression [22,23], while others found that cannabis users and nonusers were equally prone to develop depression [24,25]. Another longitudinal study suggests that MDD is associated with future initiation of cannabis use, hence suggesting self-medication [15].
Overall, it seems that depressed individuals may start using cannabis or increase the frequency of cannabis use as a way to “self-medicate” and relieve their symptoms; on the other hand, cannabis use may increase the risk for depression in heavy users who initiated their consumption in early adolescence [26,27]. Cannabis users who initiated early use and frequently used cannabis during adolescence might be at risk to develop cannabis use disorder (CUD) [28]. The estimated chances of becoming addicted to cannabis after lifetime exposure are 8.9% [29].
Cannabinoids are molecules that act on cannabinoid receptors type 1 and 2 (CB1r and CB2r) and can be divided into three broad categories: endogenous cannabinoids, synthetic cannabinoids, and plant-derived cannabinoids. The main endogenous cannabinoids are the signaling lipids N-arachidonoylethanolamine (anandamide, AEA) and 2-arachidonoylglycerol (2-AG). Synthetic cannabinoids are produced by academic laboratories or the pharmaceutical industry for research (e.g., HU-210, WIN 55,212-2, CP 47,497) or produced as popular recreational drugs [30,31]. They have a pharmacological effect by binding to CB1r and/or CB2r, with CB1 agonists responsible for the recreational effects of the synthetic cannabinoids; their effects are considered to be intense and faster than those observed with cannabis smoking, explained partly by the full agonist activity and high affinity for cannabinoid receptors [30]. The cannabis plant contains over 500 constituents, with the main compounds including delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) [32]. THC is the main psychoactive compound in cannabis, which produces the “high” sensation and could lead to adverse consequences. CBD that is derived directly from the hemp plant exhibits no effects indicative of any abuse or dependence potential in humans [33]. The cannabis plant also contains chemicals such as alkaloids, terpenes, flavonoids, phenolic acids, etc., that may elicit physiological responses in humans, some of them anxiolytic [34].
The effects of the cannabis plant have been studied in both humans and rodents in order to elucidate the involvement of the ECS in the etiology and treatment of psychiatric disorders (for recent reviews: [35,36,37]). Furthermore, positive effects have been reported when using whole plant extracts, where the whole spectrum of cannabinoids and other bioactive and non-active compounds is present; this is called the “entourage effect” [38].
Human studies for treating different psychiatric disorders have mostly focused on the cannabis sativa plant and its main compounds THC, CBD or a combination of them [17]. In studies using rodent models, researchers are attempting to isolate new cannabinoid agonists to examine their potential therapeutic effects [39,40,41,42].
When considering the efficacy of cannabis and cannabinoids for depression (or any other neuropsychiatric condition), it should be taken into consideration that cannabis has multiple components. There is a high diversity across types and strains of herbal cannabis, and pharmacological differences across cannabinoids, but only a few studies in humans that have compared these differences [43]. Hence, assessing the relative effectiveness of different cannabis strains and different cannabinoids for diverse outcomes requires further research.
In this article, we will provide an overview of the neuromodulatory effects of cannabinoid compounds on different components of the ECS (such as receptors and ligands). While studies on the effects of the whole plant on depression are important, examining the differential effects of cannabinoids on ECS components may improve our diagnosis and enhance our treatment options. To that end, we review the current knowledge about the role played by the various components of the ECS in the etiology and treatment of depression in animal models and in humans.
In the following sections, we briefly describe the ECS and then review the effects of cannabinoids on ECS components in rodent models of specific depression-like behaviors and endophenotypes. We will review the literature about the effects of agonists and antagonists of cannabinoid receptors and then discuss the findings regarding CBD. Next, we describe the findings in human subjects, specifically, in subjects with a primary diagnosis that is not depression and in self-medicating subjects. Then we will review studies assessing alterations in ECS components in depressed patients (in endogenous ligands and cannabinoid receptors) and genetic variants of the ECS that are associated with depression.

2. Cannabis and the Endocannabinoid System (ECS)

Cannabis is the most commonly used addictive substance following tobacco and alcohol, and the number of cannabis users continues to increase [44,45]. Each strain of the cannabis plant consists of roughly 120 phytocannabinoids, the most studied of which are CBD and THC [46]. Much of the interaction of these phytocannabinoids with the mammalian nervous system is through the ECS. The main receptors of the ECS are CB1r and CB2r, cannabinoid receptors belonging to the category of G-protein-coupled receptors. CB1r is found primarily in the brain and CB2r is expressed mainly in peripheral organs, especially cells associated with the immune system [47], though it is also present in the brain [48]. Other non-CB1r/non-CB2r targets of cannabinoids include transient receptor potential vanilloid 1 (TRPV1), G-protein-coupled receptor 55 (GPR55), and peroxisome proliferator-activated receptors (PPARs). As mentioned above, the main endogenous ligands of CB1r and CB2r are AEA and 2-AG. AEA is a high-affinity, partial agonist of CB1r that is almost inactive at CB2r; 2-AG is a full agonist of both CB1r and CB2r, with moderate-to-low affinity; AEA and 2-AG also interact with TRPV1 and GPR55 [49,50,51].
AEA and 2-AG are produced at postsynaptic neurons and are lipophilic molecules that are synthesized “on demand” from membrane phospholipids. They are released immediately and without being stored in vesicles. The enzymes responsible for degrading AEA and 2-AG are, respectively, the fatty acid amide hydrolase (FAAH) and the monoacylglycerol lipase (MAGL). AEA is hydrolyzed in postsynaptic neurons by FAAH, thus terminating the AEA action at the time of its synthesis, whereas 2-AG is hydrolyzed in presynaptic neurons by MAGL, following CB1r activation.
CBD acts on several targets, including the serotonin 1A receptor (5-HT1A), GPR55 and TRPV1 [52,53], as well as CB1r and CB2r, with low affinity [54]. It is a GPR55-antagonist [53] and acts as a negative allosteric modulator of CB1r, modifying the power and efficiency with which endogenous cannabinoids activate the receptor [55], and as an inverse agonist at very high concentrations [56]. CBD also inhibits the metabolization of FAAH, which increases AEA tone; it has been suggested that this is the mechanism by which CBD activates CB1r [57]. THC is an agonist of both CB1r and CB2r, with lower affinity than several other synthetic cannabinoids such as WIN55,212-2, CP55940, and the endocannabinoid 2-AG, but with similar affinity as AEA [58]. The characterization of the mode of action of THC underlies a wide spectrum of pharmacological effects, which encompass euphoria, calmness, appetite stimulation, sensory alterations, and analgesia [59].

3. Studies of Depression in Rodent Models

Rodent models for depression do not represent the human condition in its entirety [60]; rather, they represent specific features of depression. In this way, they achieve a better understanding of one critical biological function of the disease to help translate it to the human condition [61,62]. They also provide a crucial approach to examine neural circuitry and molecular and cellular pathways in a controlled environment.
Widely used models for depression are chronic mild stress (CMS) or chronic unpredictable mild stress (CUMS), chronic social defeat stress, learned helplessness, and early life stress (ELS); all cause significant changes in behavior, brain functioning and physiology. CMS/CUMS comprises a series of trials, such as day and night reversal, tail clipping, and water or food deprivation, for a period of 3 weeks or more. The model uses repeated stressors to avoid the stress adaptation that may occur following a single repeated stimulation. The chronic social defeat stress comprises of repeatedly exposing naïve male mice to aggressor mice. In learned helplessness, animals manifest a low intention to escape in an environment of uncontrollable and unpredictable injury stimulation. In ELS, adverse events in early life substantially affect the development of psychiatric illnesses in late life, such as depression [63,64].
The behavioral outcome measured in rodents is usually despair-like behavior and anhedonia. Anhedonia, a loss of interest in things that were once pleasurable, is a common symptom of depression as well as other mental health disorders. These tests are also used to measure the antidepressant potential of new compounds. In the forced swim test (FST) and the tail suspension test (TST), a rodent is exposed to a stressful and inescapable situation (swimming in the FST and suspension by its tail in the TST), and the duration of its immobility is measured. The FST is based on the assumption that when placing an animal in a container filled with water, it will first make efforts to escape but eventually will exhibit immobility that may be considered to reflect a measure of behavioral despair. These tests have good predictive validity and are able to identify drugs that may be effective in depressed patients [65]. Another frequently used measure is the saccharine/sucrose preference test: the consumption of sweetened water or choosing between sweetened water and plain water in order to measure sensitivity to reward. Decreased consumption of palatable solutions or decreased preference are considered to reflect the condition of anhedonia [66].
Most preclinical research on depression has been performed on male rodents [67]. This is despite the fact that, in humans, depression is more prevalent in women than men [68]. Furthermore, men and women in most cases differ at baseline and in their responses to stress and drugs [60], which emphasizes the need to study both sexes.

4. The Effects of Cannabinoids on ECS Components in Rodents

4.1. CB1r

CB1r in the central nervous system is distributed densely in limbic regions associated with stress and cognition, including the nucleus accumbens (NAc), hippocampus, amygdala, and paraventricular nucleus (PVN) of the hypothalamus. CB1r is abundant in the prefrontal cortex (PFC), as well as in areas involved in pain transmission and modulation; in motor regions such as the basal ganglia and cerebellum; and in glial cells and the periphery [69,70]. In this section, we review the effects of cannabinoids on CB1r in rodent models for depression.

4.1.1. Pre-Clinical Studies of CB1r Knockout and Antagonism

A number of studies have indicated a major role for CB1r in the etiology of depression, and it is estimated that its intact function is essential for a healthy mood [71]. Several studies have shown that CB1-knockout or knockdown-mice are prone to depressive-like behavior [72,73]. For example, CB1-knockout mice that were exposed to CUMS exhibited an augmented susceptibility to develop an anhedonic state, suggesting increased depressive-like behavior [73]. In a more recent study, exposure to the chronic social defeat model selectively potentiated excitatory transmission in cholecystokinin glutamatergic neurons in the basolateral amygdala (BLA) and D2 medium spiny neurons in the NAc via reduction of presynaptic CB1r [74]. Importantly, knockdown of CB1r in this circuit increased stress susceptibility, and the CB1r agonist administered to the NAc had antidepressant-like effects. This suggests that downregulating CB1r in this circuit is essential for stress-induced depression [74].
Chronic CB1r-antagonists can also result in a depressed mood. For example, 21-day intraperitoneal (i.p.) treatment with the CB1r-antagonist rimonabant (10 mg/kg) increased immobility time in the FST (i.e., elevated levels of despair-like behavior) and decreased sucrose preference (i.e., anhedonia) [75]. A summary of the effects of CB1 antagonists on depression-like behavior in rodents is presented in Table 1.
In addition, acute or chronic AM251 administration (0.3, 1 mg/kg, i.p.) to rodents exposed to stress-induced depression can inhibit the antidepressant-like effects induced by other substances and methods, such as AEA [83], repetitive transcranial magnetic stimulation [84], the synthetic non-selective cannabinoid receptor agonist WIN55,212-2 [85], the MAGL inhibitor JZL184 [86], CBD [87], the FAAH inhibitor URB597 [86] and the AEA reuptake inhibitor AM404 [88]. Acute administration of rimonabant (3 mg/kg, i.p.) prevented a decrease in immobility time in the FST induced by URB597, AM404 and CP55,940 [89]. URB597 inhibits AEA degradation and enhances AEA availability in the synapses, and thus functions as an indirect agonist of CB1r. AM404 is another enhancer of AEA, as it acts as an AEA reuptake inhibitor. CP55,940 is a potent and non-selective synthetic cannabinoid agonist. A summary of the effects of CB1 antagonists co-administered with cannabinoid agonists on depression-like behavior in rodents is presented in Table 2.
Intracerebral injections of AM251 showed similar effects; AM251 (0.8 μg) microinjection to the NAc inhibited antidepressant-like effects induced by the antidepressant phenylglycine derivative (RS)-2-chloro-5-hydroxyphenylglycine in mice that underwent the chronic social defeat stress [90]; intracerebroventricular (i.c.v.) injection of AM251 (1 μg) prevented anti-depressant effects induced by URB597 [76]. AM251 (0.28 ng) microinjection to the PFC augmented depressive-like behaviors induced by CUMS [77] and prevented the therapeutic-like effect of URB597, which decreased immobility time in the FST [91]. AM251 (0.01 ng) to the CA1 region of the hippocampus induced despair-like behavior in the FST [78,79].
Even though the majority of research shows that CB1r-antagonists enhance depressive-like behaviors, several studies have found the opposite; two-time oral administration of rimonabant (3 and 10 mg/kg) reduced immobility time in the FST in naive mice [82], suggesting an antidepressant effect; chronic oral administration of rimonabant (10 mg/kg) for 5 weeks reduced immobility time in CMS mice [82]; acute AM251 (0.3, 0.5, 1 and 10 mg/kg) reduced immobility time in the FST in mice [80] and 0.3, 0.5 and 1 mg/kg decreased immobility in the TST [80]. AM251 (0.25 mg/kg, i.p.) also augmented the antidepressant effects of tianeptine (a tricyclic antidepressant) and agomelatine (an atypical antidepressant) in mice [92]. In addition, intra-BLA microinjection of AM251 (0.01μg/ 0.5μL) reduced immobility time in the FST in rats [81].
It is interesting that rimonabant had opposite effects when administered orally, compared to i.p. and microinjections [75,82]. This suggests that different mechanisms may mediate its effect when ingested and not injected systemically. As for AM251, acute i.p. administration decreased depression-like behavior [82]. However, when administered following AEA treatment, acute AM251 blocked the antidepressant effects induced by AEA [83]; similarly, when chronically co-administered with other treatments (e.g., CB1r agonists), it blocked their therapeutic-like effects on behavior [79,80,84,85,86]. This emphasizes the complex mechanisms underlying the effects of CB1r-antagonists on depression. This complexity is further stressed by the dose-dependent, biphasic effects of CB1 ligation found in multiple studies regarding different effects on the ECS [93,94,95,96].
Taken together, these results propose that CB1r deficiency represents a model for depressive-like disorders [97], but the diversity of these findings suggests that more study is needed to fully understand the role played by CB1r-antagonism in depression.

4.1.2. Pre-Clinical Studies of CB1 Receptor Agonism

AEA generally has antidepressant properties. Multiple studies have shown that chronic i.p. injection of the FAAH-inhibitor URB597 (0.2, 0.3, 0.4, 5.8 mg/kg), which increases AEA levels, prevents depressive-like behaviors induced by different models and methods, such as CUMS [98], adolescent THC exposure [99], CMS [100] and chronic constriction injury (CCI) that induces neuropathic pain and depression-associated behavior [101]. We showed that 14-day administration of URB597 (0.4 mg/kg, i.p.), the MAGL inhibitor JZL184 (2 mg/kg; i.p) or the CB1/2 receptor agonist WIN55,212-2 (1.2 mg/kg, i.p.) during late adolescence decreased depressive-like behaviors induced by ELS in male and female rats [86,102,103]. However, when administered at mid-adolescence, the same dose of URB597 did not prevent the deleterious long-term effects of ELS exposure on depression-like behavior in males and females and induced long-term, depressive-like behavior by itself in non-stressed rats [103]. This suggests that URB597 may have deleterious or ameliorating effects on behavior, depending on the developmental time window of treatment (i.e., mid- or late adolescence) [103].
Both chronic and acute URB597 (0.3 mg/kg) and AM404 (5 mg/kg) prevented depressive-like behaviors in the FST induced by severe electric shock [40] and nicotine abstinence [88], respectively. The same effect was seen in naive rats, in which URB597 (0.1, 0.3, 1, 3.2 mg/kg, i.p.), AM404 (0.3, 1, 3 mg/kg, i.p.), CP55,940 (0.1 mg/kg, i.p.), and the CB1r-agonist oleamide (10, 20 mg/kg, i.p.) decreased FST immobility time, supporting the antidepressant-like effects of these compounds [89,104,105,106]. Oleamide, a fatty amide derived from oleic acid (5 mg/kg, i.p.), also augmented the antidepressant-like effects of the atypical antidepressant tianeptine in the FST [78]. The CB1r synthetic agonist arachidonyl-2′-chloroethylamide (ACEA; 10 mg/kg, i.p.) increased sucrose consumption in post-stroke depression rats, suggesting decreased anhedonia [107]; post-stroke depression is one of the most common psychological consequences of stroke.
I.c.v. injection of URB597 (5 and 10 ng) prevented depressive behaviors that were induced by methamphetamine in mice [76]. Despair-like behavior also was decreased in mice by i.c.v. administration of URB597 (0.05, 0.1, 1, 5, 10 μg), AM404 (0.1, 1, 5, 10 μg), and AEA (1, 5, 10, 20 μg) [108], and by microinjection of URB597 (0.01, 0.1, 1 nmol) to the ventromedial PFC [109]. Microinjection of URB597 (0.01 μg) to the PFC in rats reduced FST immobility time [91]. No effect on FST performance, however, was seen after microinjection of URB597 (0.5, 1 μg) to the dentate gyrus of the hippocampus. However, administration of the CB1/CB2-agonist HU-210 (1, 2.5 μg) to the same region decreased immobility time in the FST [110]. A summary of CB1-mediated effects of cannabinoids on depression-like behavior in rodents is presented in Table 3.
All things considered, the data propose that augmenting ECS signaling via CB1r may be a novel approach to decrease depression-like behavior and that the use of CB1r antagonists warrants caution. Specifically, FAAH inhibition, which enhances AEA-mediated CB1r signaling, has been suggested to generate a more specific and beneficial spectrum of biological effects than those caused by direct CB1r agonists [111,112].

4.2. CB2r

CB2r was discovered at the beginning of the 1990s, and at first, it was assumed to be present mainly in peripheral and immune tissues [113]. However, its presence has been observed in some subsets of neurons in the brain and thus this receptor likely participates in the modulation of neurotransmission [114]. CB2r is mainly studied in pain and inflammation, yet a growing number of studies provide evidence of a potential role of CB2r in the etiology of depression [115]; the main endogenous ligand for CB2r is 2-AG [116].

4.2.1. Pre-Clinical Studies of CB2 Knockout and Antagonism

The outcomes of CB2-antagonists administration seem to be dose-dependent. On the one hand, it may enhance depressive-like behaviors, or invert the antidepressant effects of CB2r-dependent treatments. On the other hand, it may facilitate antidepressant effects induced by other treatments. For example, the CB2-inverse agonist AM630 (1 mg/kg i.p.) blocks the antidepressant effects in the FST induced by CBD in diabetic rats [87], but works as an antidepressant when administered at a lower dose (0.5 mg/kg, i.p.) [104]; diabetic patients are two to three times more likely to develop depression and diabetic rats demonstrate depression-like behaviors [117]. When administered acutely at a low dose (0.25 mg/kg, i.p.) in mice, AM630 augments the antidepressant effects of the tricyclic antidepressant imipramine, the SSRI escitalopram, the norepinephrine reuptake inhibitor reboxetine, and the atypical antidepressants agomelatine and tianeptine [92,118].

4.2.2. Pre-Clinical Studies of CB2 Agonism

Several studies report that CB2r agonists have antidepressant properties. For example, the CB2-full agonist β-Caryophyllene (BCP) ameliorated depressive-like behaviors (i.e., reduced immobility time in the TST and FST) when acutely administered i.p. (50 mg/kg) in mice [119] and chronically administered (25, 50, 100 mg/kg) in rats that were subjected to daily restraint stress [120]. Chronic oral administration of BCP (10 mg/kg) was effective in reducing immobility in the TST in diabetic mice [121].
An acute low dose of the CB2 agonist JWH 133 (0.25 mg/kg, i.p.), increased the antidepressant effects of the tricyclic antidepressant imipramine, the SSRI escitalopram and the norepinephrine reuptake inhibitor reboxetine in mice [122], while higher doses (0.5, 1 mg/kg, i.p.) had similar effects on their own [104]. JWH133 significantly decreased anhedonia (i.e., increased sucrose consumption) when injected i.p. (5 mg/kg) for 7 days or when microinjected (3 μg) acutely into the ventromedial hypothalamus of post-stroke depression rats [107].
The CB2-agonist GW 405833 has been mainly studied as a treatment for pain, and was found to reverse depressive-like behaviors induced by chronic constriction injury (CCI) in rats (30 mg/kg, i.p.) [122]. Another study found that CB2 agonists and overexpression of CB2 were correlated with decreased depressive-like behavior in transgenic mice, as evidenced by the FST and the novelty-suppressed feeding test [123]; the novelty-suppressed feeding test is sensitive to chronic, but not acute, antidepressant treatment, and is assumed to mirror the effects of antidepressant treatment in human patients.
Overall, compounds used to activate CB2r seem to intensify the antidepressant-like effects induced by other drugs. This suggests that CB2r is involved in depression-related behaviors through interactions with other systems that modulate these responses (e.g., serotonergic).

4.3. GPR55

The GPR55 receptor was cloned in 1999 [124] and was later characterized as part of the ECS, as it binds AEA and 2-AG as well as THC and CBD [50,51]. There is evidence that GPR55 plays an important role in depression; a 7-day intravenous (i.v.) treatment with the GPR55-agonist O-1602 decreased despair-like behavior in female rats subjected to a 14-day corticosterone treatment [125]. A 10-day chronic social defeat stress lowered hippocampal GPR55 levels in mice that were susceptible to the model (i.e., showed elevated levels of depression and anxiety), but not in resilient mice. Interestingly, O-1602 treatment (10 mg/kg, i.p.) during chronic social defeat stress decreased these behaviors [126]. Compared to a control group, the learned helplessness model decreased GPR55 mRNA levels in the lateral habenula and the amygdala, with no effects in the hippocampus and medial PFC [127]. To summarize, these studies suggest that exposure to stress-induced depression results in decreased levels of GPR55 in a region-dependent manner and that the GPR55 agonist has antidepressant effects on behavior.

4.4. TRPV1

TRPV1 is a nociceptive receptor that has been thoroughly studied in the context of pain [128]. Considering the large comorbidity of pain and depression [129], it is not surprising that there are interesting findings regarding the role of TRPV1 in depression. It is generally assumed that TRPV1-agonists induce depressive behavior, and that TRPV1-antagonists may provoke the opposite effect [130].
Three injections (2.5, 5 mg/kg, i.p.) of AA-5-HT (a dual blocker of FAAH and TRPV1) reduced immobility time in stressed rats [131]. AA-5-HT attenuated despair-like behavior in rats, also when microinjected (0.25, 0.5 nmol) into the PFC [109,132]. TRPV1 mRNA levels in the medial PFC of mice that underwent the learned helplessness model of depression were significantly lower than in control mice [127].

4.5. CBD

CBD has multiple key targets, including cannabinoid receptors, 5-HT1A receptors, and neurogenesis factors, and hence is addressed separately in this section. Studies have shown its potential to treat depressive-like behaviors; for example, acute treatment with CBD (200 mg/kg, i.p.) reduced immobility time in the FST in mice [133]; a 7-day treatment in adolescent rats, adult rats and mice (10, 30, 100 mg/kg, i.p., respectively) reduced FST immobility time [134,135]. Moreover, a sub-chronic administration of CBD (30 mg/kg, i.p.) reduced despair-like behavior in diabetic mice [136]; a chronic, 28-day administration of CBD (10 mg/kg, i.p.) elevated sucrose intake in CUMS rats [137]. CBD was effective in lowering depression behaviors in two rat strains genetically modified for depression research; acute oral CBD (30 mg/kg) reduced immobility time in the FST and elevated saccharine consumption in male and female Wistar Kyoto rats and reduced immobility in males of the Flinders Sensitive Line [39,138].
Acute CBD (10 mg/kg, i.p.) lowered FST immobility time in mice, both 30 min and 7 days after administration [139,140,141]. A lower dose of CBD (7 mg/kg, i.p.) was as effective in lowering FST immobility time when co-administered with ineffective doses of the TCA desipramine, the SSRI fluoxetine, and the DNA-methylation inhibitors AzaD and RG108 [139,141]. The short-term antidepressant effects of CBD were associated with increased medial PFC expression of synaptophysin, PSD95, and brain-derived neurotrophic factor (BDNF), as well as elevated hippocampal BDNF [142]. Chronic CBD (15 mg/kg, i.p.) treatment also increased BDNF levels in the amygdala; a higher dose of CBD (30 mg/kg, i.p.) produced antidepressant-like effects in the FST when administered acutely and chronically [142]. Both lower (10 mg/kg, i.v.) and higher doses (100 mg/kg, oral) had antidepressant properties in CMS mice, in association with increased BDNF and synaptophysin mRNA in the medial PFC and hippocampus [143]. In addition, microinjection of CBD (15, 30, 60 nmol) to the pre-limbic division of the medial PFC of neuropathic pain-mice resulted in lower despair-like behavior in the FST [144].
Although CBD interacts with many ECS receptors (including those for CB1, CB2, TRPV1, and GPR55), studies of its antidepressant properties have focused mainly on the serotonergic 5-HT1A receptor as the main receptor that mediates these effects: Acute CBD (30 mg/kg, i.p.) reduced immobility in the FST in naive mice, an effect that was blocked by the 5HT1A-antagonist WAY100635 [145]; 7-day administration of CBD (50 mg/kg, i.p.) improved sucrose intake in mice that underwent the olfactory bulbectomy model of depression; both antidepressant-like effects and enhanced cortical 5-HT/glutamate neurotransmission induced by CBD were prevented by 5-HT1A receptor blockade [146]; similarly, microinjection of CBD to the ventromedial PFC (10, 30, 60 nmol to the prelimbic subregion; 45, 60 nmol to the infralimbic subregion) reduced FST immobility time, an effect that was blocked by pretreatment with WAY100635; the CB1-antagonist AM251 also blocked the effects of CBD [147]. Interestingly, a study of the anxiolytic effects of chronic CBD (30 mg/kg, i.p.) identified a mediating role for CB1r and CB2r but not for 5HT1A receptor. Taken together, these studies suggest that the effects of CBD on anxiety and depression may be mediated by different mechanisms [148]. A summary of the effects of CBD on depression-like behavior in rodents is presented in Table 4.

5. The ECS in Human Studies of Depression

There is accumulating preclinical evidence that targeting the ECS could potentially benefit patients suffering from depression [149]. However, epidemiological and clinical studies do not provide strong evidence to support that cannabis can be used as an antidepressant [150].
In reviewing the literature, we found very few studies where cannabinoids were used to treat depression with well-designed, randomized control trials (RCTs). Yet there are other strong indications from human studies that encourage further research. The potential role for the ECS in depression comes from a series of studies indicating that the CB1r antagonist rimonabant is associated with the development of severe adverse effects, including depression and suicide [151]. Clinical observations showed that cannabis stimulates appetite (the “munchies”) [152]. Rimonabant was developed as an anti-obesity treatment. A meta-analysis conducted in 2007 concluded that 20 mg/day of rimonabant increases the risk of depressive symptoms [153]. A later study, however, found that rimonabant in the same dosage had no effect on mood [154]. These findings are in line with an FDA report about the safety of rimonabant, which stated that 26% of the subjects given 20 mg rimonabant daily later developed psychiatric symptoms, compared to 14% of those given placebo [154].

5.1. Subjects with a Primary Diagnosis That Is Not Depression

There are no published RCTs that examined the direct effect of THC or CBD on depressive symptoms [155]. However, many published RCTs have examined the effects of THC or THC:CBD on other conditions, such as pain and multiple sclerosis (MS) [156]. These RCTs did not find improvement in depression symptoms in these patients compared to placebo. For example, Nabiximols (Sativex; an equal mix of THC and CBD) produced no effect on symptoms of depression in people with MS or with chronic pain due to cancer [157,158]. A small RCT that examined the effects of CBD alone in chronic pain, also found no change in depressive symptoms [159].
It is important to note that these RCTs did not include subjects with a primary diagnosis of depression (i.e., depression was assessed indirectly); that the patients’ self-reported depression scores were already low [160,161,162]; and that psychiatric diagnosis was listed as an exclusion criterion in some of the studies [157,158]. In total, this makes it difficult to extrapolate these outcomes to people with clinical depression.
However, there are studies that support an antidepressant effect of cannabinoids when depression was assessed directly, although not the primary diagnosis. In one study, a cross-sectional, longitudinal, and experimental design on individuals with social pain showed that marijuana use predicted lower levels of later depression among participants who were lonely, and that those individuals who used marijuana relatively frequently were less likely to have experienced a DSM-IV major depressive event during the previous 12 months [163].
A similar result was observed in a study that examined the effects of prolonged CBD administration on psychological symptoms and cognition to a community sample of regular cannabis users; oral CBD reduced depressive- and psychotic-like symptoms and improved attentional switching, verbal learning, and memory. Moreover, CBD was well tolerated with no reported side effects [164]. Similarly, in a randomized, double-blind, inpatient trial, nabiximols was used as an agonist therapy for reducing the severity and time course of cannabis withdrawal and for retaining participants in withdrawal treatment; nabiximols suppressed withdrawal-related irritability, cravings, and significantly reduced depression [165].

5.2. Self-Medication Studies

There is evidence that pharmacological interventions (specifically, SSRIs) may be effective in treating depression at the population level, but may not always be visible at the patient level [155]. The small effect size of SSRIs [17] together with their adverse side effects, means that some depressed individuals often seek alternative treatments. As a result, patients with depression are increasingly using medicinal cannabis products to relieve their symptoms [155].
Some reports of depressed patients self-medicating with cannabis demonstrate lower levels of depressive symptoms and improved sleep [166,167,168]. However, other reports show adverse effects, as depressed patients who self-medicate with cannabis demonstrate increased mental health problems and lowered improvement in depression symptoms and suicidal ideation [169].
In a longitudinal, cross-sectional study, medicinal cannabis users reported reduced depression and improved quality of life compared to a control group that was considering (but had not yet initiated) medicinal cannabis use [170]. In an observational study, medicinal cannabis use reduced depressive symptoms in clinically depressed populations [155]; specifically, medicinal cannabis use was associated with better sleep, quality of life, and less pain. Moreover, the group that initiated cannabis use during the follow-up period demonstrated fewer depressive symptoms compared to a control group that never initiated cannabis use [155].
To correctly interpret the therapeutic potential of cannabis, it is important to restrict and separate the cannabis effects reported under recreational consumption compared to clinical trials under medical supervision. Although many people report using cannabis to manage a large variety of medical conditions, including depression, the gathering of information regarding self-medication makes it hard to draw firm quantitative conclusions about the effectiveness of treatment.

5.3. ECS Components Altered in Depression

Compelling evidence for the involvement of the ECS in depression comes from studies assessing alterations in ECS components in depressed patients. Elucidating the effects of depression on different targets of the ECS is important because the ECS is involved in eliciting potent effects on neurotransmission, neuroendocrine, and inflammatory processes, which are known to be disturbed in depression.

5.3.1. Endogenous Ligands

Accumulating data suggest that depression is strongly associated with deficient endocannabinoid signaling [171], and hence provide compelling evidence for the involvement of the ECS in the etiology of depression and a rationale for activating the ECS to relieve depression. For example, serum levels of AEA and 2-AG were found to be decreased in depressed patients [172]. Interestingly, the reduction in 2-AG serum levels was negatively correlated with the duration of depressive episodes [173]. Another study found indications of a deficit in peripheral endocannabinoid activity: basal serum concentrations of AEA and 2-AG were significantly decreased in women with MDD relative to matched controls [172]. Other studies have shown increased levels of endocannabinoids following treatment; plasma levels of oleoylethanolamide, AEA, and 2-AG were increased in patients with depression treated with SSRIs compared to a non-depressed control group [174]. Similarly, in men and women patients with MDD, physical exercise elevated plasma levels of AEA and 2-AG; the authors suggested that endocannabinoids may contribute to the antidepressant effects of exercise in MDD [175,176]. Antidepressant treatment by electroconvulsive therapy (ECT) elevated AEA and to some extent 2-AG levels in the cerebrospinal fluid [177]. These studies indicate that the ECS is modulated by effective antidepressant treatment.

5.3.2. CB1r

Studies have found indications of increased CB1r availability in depression. Concentrations of CB1r and CB1rmediated stimulation of G proteins in the PFC were found to be increased in subjects with major depression who had died by suicides relative to controls [178,179]. Similarly, treatment with SSRIs decreased expression of CB1r in the anterior cingulate cortex of postmortem MDD patients [180]. However, another study did not find changes in CB1r protein expression in depressive subjects compared to controls [173]. Increased CB1r availability in depression may be a compensation response to low AEA levels, as suggested in post-traumatic stress disorder (PTSD) [112].
It should be noted that chronic direct activation of CB1r downregulated CB1r [181,182], which may in turn result in a depression-like phenotype in certain individuals [75,183].
Taken together, the data suggest that enhancing endocannabinoid signaling may serve as an antidepressant; CB1r blockade produces depression; and chronic direct activation of CB1r may produce region-dependent CB1r desensitization and down-regulation that is associated with depression [21].

5.4. Genetic Studies

Several studies have reported associations between genetic variants of the cannabinoid receptor type 1 and type 2 genes (CNRs; CNR1 and CNR2) and a susceptibility to develop depression. However, such studies reported conflicting findings [184]. Genetic variants of CNRs can affect gene transcription (and, thereby, protein expression and biologic function) of these cannabinoid receptors [185]. ECS-related polymorphic gene variant alterations have been reported, which may have both diagnostic and therapeutic implications.

5.4.1. CNR1

The interaction between specific genetic variations in CNR1 and the vulnerability to depression has recently gained great interest. In a population of opiate-dependent outpatients remitted under stable methadone treatment, subjects with one single nucleotide polymorphism (SNP) of the CNR1 (named rs2023239) had a lower prevalence of lifetime MDD [186]. However, two other studies found no relation between CNR1 microsatellite polymorphisms and depressive disorders [187] or between CNR1rs1049353 and MDD [188,189]. Another piece of evidence against the relationship between depression and CNR1 is a recent meta-analysis, which assessed the relation between CNR1 and CNR2 polymorphisms and depressive disorder susceptibility. This meta-analysis did not find a significant association of the CNR1rs1049353 SNP with depressive disorders [184].

5.4.2. CNR2

Some studies indicate that dysfunctional CB2r can contribute to greater sensitivity to childhood trauma, a risk factor for depression; specifically, the CNR2 R63Q polymorphism was associated with anxious and depressive phenotypes following childhood trauma [190]. In support, the expression of CNR2 Q63R was also found to be higher in Japanese depressed patients [191] and alcoholics [192] (alcoholism is in high comorbidity with MDD [193]). One study reported a higher incidence of the CB2 allele Leu133Ile for bipolar disorder patients [194]. A recent meta-analysis found a significant association of CNR2rs2501432 with depressive disorders [184]; also, in the dorsolateral PFC of suicide victims, CB2 gene expression was 33% lower, but their levels of CB2 protein were higher, when compared to a control group. This difference might stem from a compensatory mechanism that controls gene half-life and protein turnover [195].
To summarize, genetic variations in CNR2 are associated with the vulnerability to depression; relating this marker to depression-associated brain dysfunction may potentially improve the diagnosis and treatment for depression.

5.4.3. FAAH

In the aforementioned study that showed an association between CNR2 R63Q polymorphism and depression, the researchers also found that dysfunctional FAAH could contribute to greater sensitivity to childhood trauma [190]; specifically, the FAAH rs324420 polymorphism (i.e., C385A) was associated with anxious and depressive phenotypes following childhood trauma [196]. The same polymorphism was higher in MDD patients and bipolar disorder patients [189]. In a study in cannabis users, greater past-year cannabis use and FAAH rs324420 genotype predicted poor sleep quality, which was mediated by depressive symptoms. Moreover, participants with higher cannabis use and depressive symptoms reported more impaired sleep [197].

5.4.4. GPR55

In a study that evaluated alterations of GPR55 in suicide victims compared to corresponding controls, GPR55 gene expression was 41% lower in the dorsolateral PFC of suicide victims, and GPR55 protein in these subjects were the same as in the control group [195]. The link between suicide and mental disorders (in particular, depression and alcohol use disorders) is well established, and dysfunctions in the dorsolateral PFC of patients who attempted suicide are associated with impaired executive functions and increased impulsivity [198].
To summarize, evidence from longitudinal studies suggests that depression might increase cannabis use and perhaps vice versa. There is evidence of alterations to the genetic and ECS components in depression, suggesting that the ECS may be critically involved in the pathophysiology of depression.

5.4.5. Caveats

There is a growing belief that cannabis and other cannabinoids are harmless drugs that can decrease anxiety and depression and induce relaxation. Accordingly, the use of medicinal cannabis and cannabinoids has recently been increasing. However, only a few registered drugs (usually containing CBD and THC) are of high quality [35]. In fact, in many of the above studies, the sources of cannabis are unknown or uncontrolled [199]. Moreover, several studies showed that the effects of cannabis on depression symptoms may be positive or negative, depending on the time course of administration; hence, although it was found that cannabis provided a brief relief, the long-term effects were worsening of symptoms [200]. This suggests that the short- and long-term effects of cannabis in depression should be taken under consideration. Additionally, we should bear in mind the potential risk for adverse events during cannabinoid usage. There are reports of increased risk of acute psychotic symptoms [201], and in young adults, chronic daily use of cannabis might generate cannabis dependence [202].

6. Conclusions

Rodent studies strongly suggest that activating the ECS produces antidepressant-like responses in a variety of behavioral tests. The effects are dependent on dosing, route of administration, and other factors, but the overall effect is that both direct and indirect activation of ECS components and CB1r in particular have an antidepressant potential, whereas deficits in ECS signaling may have depressive effects.
In humans, most studies addressed a different primary medical disorder (pain, MS) other than depression, with depression as a secondary condition. In these studies, cannabinoids had no effect on depression [17,150], but there is a lack of high-quality studies where depression is the primary target of cannabis treatment. Studies that examined patients with a primary diagnosis of depression included a small sample size and other methodological flaws [150]. In addition, most studies in human subjects did not compare the efficacy of cannabinoids with those of existing antidepressant agents. Therefore, high quality, large-scale RCTs in depressed patients are needed to assess the effectiveness and safety of cannabinoids and to compare it with placebo and standard treatments.
To conclude, the findings on the effectiveness of cannabis and cannabinoid compounds in depression reveal inconsistencies in the outcomes obtained in animal models compared to findings in depressed patients. By elucidating the effects of cannabinoids on ECS components, we can enhance our understanding of which targets the compounds hit, what processes they alter, and, eventually, which of these effects are needed for therapeutic efficacy.
Elucidating the role of the ECS in the etiology of depression and revealing the effects of different cannabinoids on the ECS in depression increase the probability of choosing cannabinoid compounds that will be effective treatments; this is imperative because understanding how different cannabinoid compounds work can help stratify clinical trials to focus them on those patients most likely to respond.

Author Contributions

U.B. and I.A. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by a grant from the Israel Science Foundation (ISF) (https://www.isf.org.il/#/, accessed on 29 March 2022) (992/20) to I.A.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization: Depression and Other Common Mental Disorders: Global Health Estimates. Available online: https://apps.who.int/iris/bitstream/handle/10665/254610/WHO-MSD-MER-2017.2-eng.pdf (accessed on 29 March 2022).
  2. Young, M.A.; Fogg, L.F.; Scheftner, W.A.; Keller, M.B.; Fawcett, J.A. Sex differences in the lifetime prevalence of depression: Does varying the diagnostic criteria reduce the female/male ratio? J. Affect. Disord. 1990, 18, 187–192. [Google Scholar] [CrossRef]
  3. James, S.L.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1789–1858. [Google Scholar] [CrossRef] [Green Version]
  4. Edition, F. Diagnostic and statistical manual of mental disorders. Am. Psychiatric. Assoc. 2013, 21, 591–643. [Google Scholar]
  5. Machmutow, K.; Meister, R.; Sen, A.; Kriston, L.; Watzke, B.; Härter, M.C.; Liebherz, S. Comparative effectiveness of continuation and maintenance treatments for persistent depressive disorder in adults. Cochrane Database Syst. Rev. 2019. [Google Scholar] [CrossRef]
  6. Luo, Y.; Kataoka, Y.; Ostinelli, E.G.; Cipriani, A.; Furukawa, T.A. National prescription patterns of antidepressants in the treatment of adults with major depression in the US between 1996 and 2015: A population representative survey based analysis. Front. Psychiatry 2020, 11, 35. [Google Scholar] [CrossRef]
  7. Henssler, J.; Heinz, A.; Brandt, L.; Bschor, T. Antidepressant withdrawal and rebound phenomena. Dtsch. Ärzteblatt Int. 2019, 116, 355. [Google Scholar] [CrossRef]
  8. Joshi, A. Selective serotonin re-uptake inhibitors: An overview. Psychiatr. Danub. 2018, 30 (Suppl. 7), 605–609. [Google Scholar]
  9. Montgomery, S.A.; Nielsen, R.Z.; Poulsen, L.H.; Häggström, L. A randomised, double-blind study in adults with major depressive disorder with an inadequate response to a single course of selective serotonin reuptake inhibitor or serotonin–noradrenaline reuptake inhibitor treatment switched to vortioxetine or agomelatine. Hum. Psychopharmacol. Clin. Exp. 2014, 29, 470–482. [Google Scholar]
  10. Khan, A.; Faucett, J.; Lichtenberg, P.; Kirsch, I.; Brown, W.A. A systematic review of comparative efficacy of treatments and controls for depression. PLoS ONE 2012, 7, e41778. [Google Scholar] [CrossRef] [Green Version]
  11. Cascade, E.; Kalali, A.H.; Kennedy, S.H. Real-world data on SSRI antidepressant side effects. Psychiatry (Edgmont) 2009, 6, 16. [Google Scholar]
  12. Cipriani, A.; Furukawa, T.A.; Salanti, G.; Chaimani, A.; Atkinson, L.Z.; Ogawa, Y.; Leucht, S.; Ruhe, H.G.; Turner, E.H.; Higgins, J.P.; et al. Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: A systematic review and network meta-analysis. Focus 2018, 16, 420–429. [Google Scholar] [CrossRef]
  13. Goethe, J.W.; Woolley, S.B.; Cardoni, A.A.; Woznicki, B.A.; Piez, D.A. Selective serotonin reuptake inhibitor discontinuation: Side effects and other factors that influence medication adherence. J. Clin. Psychopharmacol. 2007, 27, 451–458. [Google Scholar] [CrossRef]
  14. 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]
  15. 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]
  16. Hill, M.N.; Hillard, C.J.; Bambico, F.R.; Patel, S.; Gorzalka, B.B.; Gobbi, G. The therapeutic potential of the endocannabinoid system for the development of a novel class of antidepressants. Trends Pharmacol. Sci. 2009, 30, 484–493. [Google Scholar] [CrossRef]
  17. Sarris, J.; Sinclair, J.; Karamacoska, D.; Davidson, M.; Firth, J. Medicinal cannabis for psychiatric disorders: A clinically-focused systematic review. BMC Psychiatry 2020, 20, 24. [Google Scholar] [CrossRef] [Green Version]
  18. Keyhani, S.; Steigerwald, S.; Ishida, J.; Vali, M.; Cerdá, M.; Hasin, D.; Dollinger, C.; Yoo, S.R.; Cohen, B.E. Risks and benefits of marijuana use: A national survey of US adults. Ann. Intern. Med. 2018, 169, 282–290. [Google Scholar] [CrossRef]
  19. 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]
  20. Feingold, D.; Hoch, E.; Weinstein, A.; Hall, W. Psychological Aspects of Cannabis Use and Cannabis Use Disorder. Front. Psychiatry 2021, 12, 789197. [Google Scholar] [CrossRef]
  21. Bovasso, G.B. Cannabis abuse as a risk factor for depressive symptoms. Am. J. Psychiatry 2001, 158, 2033–2037. [Google Scholar] [CrossRef]
  22. Fergusson, D.M.; Horwood, L.J. Early onset cannabis use and psychosocial adjustment in young adults. Addiction 1997, 92, 279–296. [Google Scholar] [CrossRef]
  23. Brook, D.W.; Brook, J.S.; Zhang, C.; Cohen, P.; Whiteman, M. Drug use and the risk of major depressive disorder, alcohol dependence, and substance use disorders. Arch. Gen. Psychiatry 2002, 59, 1039–1044. [Google Scholar] [CrossRef] [Green Version]
  24. 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]
  25. Langlois, C.; Potvin, S.; Khullar, A.; Tourjman, S.V. Down and high: Reflections regarding depression and cannabis. Front. Psychiatry 2021, 12, 681. [Google Scholar] [CrossRef]
  26. Feingold, D.; Weinstein, A. Cannabis and depression. Cannabinoids Neuropsychiatr. Disord. 2021, 1264, 67–80. [Google Scholar]
  27. Mechoulam, R.; Hanuš, L.O.; Pertwee, R.; Howlett, A.C. Early phytocannabinoid chemistry to endocannabinoids and beyond. Nat. Rev. Neurosci. 2014, 15, 757–764. [Google Scholar] [CrossRef]
  28. Leung, J.; Chan, G.C.; Hides, L.; Hall, W.D. What is the prevalence and risk of cannabis use disorders among people who use cannabis? A systematic review and meta-analysis. Addict. Behav. 2020, 109, 106479. [Google Scholar] [CrossRef]
  29. Curran, H.V.; Freeman, T.P.; Mokrysz, C.; Lewis, D.A.; Morgan, C.J.; Parsons, L.H. Keep off the grass? Cannabis, cognition and addiction. Nat. Rev. Neurosci. 2016, 17, 93–306. [Google Scholar] [CrossRef]
  30. Le Boisselier, R.; Alexandre, J.; Lelong-Boulouard, V.; Debruyne, D. Focus on cannabinoids and synthetic cannabinoids. Clin. Pharmacol. Ther. 2017, 101, 220–229. [Google Scholar] [CrossRef]
  31. Papaseit Fontanet, E.; Pérez Mañá, C.; Pérez-Acevedo, A.P.; Hladun, O.; Torres-Moreno, M.C.; Muga, R.; Torrens, M.; Farré Albaladejo, M. Cannabinoids: From pot to lab. Int. J. Med. Sci. 2018, 15, 1286. [Google Scholar] [CrossRef] [Green Version]
  32. Adams, I.B.; Martin, B.R. Cannabis: Pharmacology and toxicology in animals and humans. Addiction 1996, 91, 1585–1614. [Google Scholar] [CrossRef]
  33. Hosein Farzaei, M.; Bahramsoltani, R.; Rahimi, R.; Abbasabadi, F.; Abdollahi, M. A systematic review of plant-derived natural compounds for anxiety disorders. Curr. Top. Med. Chem. 2016, 16, 1924–1942. [Google Scholar] [CrossRef]
  34. 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]
  35. García-Gutiérrez, M.S.; Navarrete, F.; Gasparyan, A.; Austrich-Olivares, A.; Sala, F.; Manzanares, J. Cannabidiol: A potential new alternative for the treatment of anxiety, depression, and psychotic disorders. Biomolecules 2020, 10, 1575. [Google Scholar] [CrossRef]
  36. Orsolini, L.; Chiappini, S.; Volpe, U.; De Berardis, D.; Latini, R.; Papanti, G.D.; Corkery, 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] [Green Version]
  37. Sbarski, B.; Akirav, I. Cannabinoids as therapeutics for PTSD. Pharmacol. Ther. 2020, 211, 107551. [Google Scholar] [CrossRef]
  38. Ben-Shabat, S.; Fride, E.; Sheskin, T.; Tamiri, T.; Rhee, M.H.; Vogel, Z.; Bisogno, T.; De Petrocellis, L.; Di Marzo, V.; Mechoulam, R. An entourage effect: Inactive endogenous fatty acid glycerol esters enhance 2-arachidonoyl-glycerol cannabinoid activity. Eur. J. Pharmacol. 1998, 353, 23–31. [Google Scholar] [CrossRef]
  39. Shoval, G.; Shbiro, L.; Hershkovitz, L.; Hazut, N.; Zalsman, G.; Mechoulam, R.; Weller, A. Prohedonic effect of cannabidiol in a rat model of depression. Neuropsychobiology 2016, 73, 123–129. [Google Scholar] [CrossRef]
  40. Burstein, O.; Shoshan, N.; Doron, R.; Akirav, I. Cannabinoids prevent depressive-like symptoms and alterations in BDNF expression in a rat model of PTSD. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 84, 129–139. [Google Scholar] [CrossRef]
  41. Hen-Shoval, D.; Amar, S.; Shbiro, L.; Smoum, R.; Haj, C.G.; Mechoulam, R.; Zalsman, G.; Weller, A.; Shoval, G. Acute oral cannabidiolic acid methyl ester reduces depression-like behavior in two genetic animal models of depression. Behav. Brain Res. 2018, 351, 1–3. [Google Scholar] [CrossRef]
  42. ElBatsh, M.M.; Moklas, M.A.; Marsden, C.A.; Kendall, D.A. Antidepressant-like effects of Δ9-tetrahydrocannabinol and rimonabant in the olfactory bulbectomised rat model of depression. Pharmacol. Biochem. Behav. 2012, 102, 357–365. [Google Scholar] [CrossRef]
  43. Huestis, M.A. Human cannabinoid pharmacokinetics. Chem. Biodivers. 2007, 4, 1770. [Google Scholar] [CrossRef] [Green Version]
  44. Degenhardt, L.; Bucello, C.; Calabria, B.; Nelson, P.; Roberts, A.; Hall, W.; Lynskey, M.; Wiessing, L.; GBD Illicit Drug Use Writing Group. What data are available on the extent of illicit drug use and dependence globally? Results of four systematic reviews. Drug Alcohol Depend. 2011, 117, 85–101. [Google Scholar] [CrossRef]
  45. United Nations: World Drug Report 2012. Available online: https://www.unodc.org/unodc/en/data-and-analysis/WDR-2012.html (accessed on 27 March 2022).
  46. Rock, E.M.; Parker, L.A. Constituents of cannabis sativa. Cannabinoids Neuropsychiatr. Disord. 2021, 1264, 1–13. [Google Scholar]
  47. Howlett, A.C. The cannabinoid receptors. Prostaglandins Other Lipid Mediat. 2002, 68, 619–631. [Google Scholar] [CrossRef]
  48. Van Sickle, M.D.; Duncan, M.; Kingsley, P.J.; Mouihate, A.; Urbani, P.; Mackie, K.; Stella, N.; Makriyannis, A.; Piomelli, D.; Davison, J.S.; et al. Identification and functional characterization of brainstem cannabinoid CB2 receptors. Science 2005, 310, 329–332. [Google Scholar] [CrossRef] [Green Version]
  49. Ross, R.A. Anandamide and vanilloid TRPV1 receptors. Br. J. Pharmacol. 2003, 140, 790–801. [Google Scholar] [CrossRef] [Green Version]
  50. Ryberg, E.; Larsson, N.; Sjögren, S.; Hjorth, S.; Hermansson, N.O.; Leonova, J.; Elebring, T.; Nilsson, K.; Drmota, T.; Greasley, P. The orphan receptor GPR55 is a novel cannabinoid receptor. Br. J. Pharmacol. 2007, 152, 1092–1101. [Google Scholar] [CrossRef]
  51. Hiley, C.R.; Kaup, S.S. GPR55 and the vascular receptors for cannabinoids. Br. J. Pharmacol. 2007, 152, 559–561. [Google Scholar] [CrossRef] [Green Version]
  52. Ibeas Bih, C.; Chen, T.; Nunn, A.V.; Bazelot, M.; Dallas, M.; Whalley, B.J. Molecular targets of cannabidiol in neurological disorders. Neurotherapeutics 2015, 12, 699–730. [Google Scholar] [CrossRef] [Green Version]
  53. Sylantyev, S.; Jensen, T.P.; Ross, R.A.; Rusakov, D.A. Cannabinoid-and lysophosphatidylinositol-sensitive receptor GPR55 boosts neurotransmitter release at central synapses. Proc. Natl. Acad. Sci. USA 2013, 110, 5193–5198. [Google Scholar] [CrossRef] [Green Version]
  54. Mechoulam, R.; Peters, M.; Murillo-Rodriguez, E.; Hanuš, L.O. Cannabidiol–recent advances. Chem. Biodivers. 2007, 4, 1678–1692. [Google Scholar] [CrossRef]
  55. Laprairie, R.B.; Bagher, A.M.; Kelly, M.E.; Denovan-Wright, E. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br. J. Pharmacol. 2015, 172, 4790–4805. [Google Scholar] [CrossRef] [Green Version]
  56. De Petrocellis, L.; Ligresti, A.; Moriello, A.S.; Allarà, M.; Bisogno, T.; Petrosino, S.; Stott, C.G.; Di Marzo, V. Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br. J. Pharmacol. 2011, 163, 1479–1494. [Google Scholar] [CrossRef] [Green Version]
  57. Stern, C.A.; da Silva, T.R.; Raymundi, A.M.; de Souza, C.P.; Hiroaki-Sato, V.A.; Kato, L.; Guimarães, F.S.; Andreatini, R.; Takahashi, R.N.; Bertoglio, L.J. Cannabidiol disrupts the consolidation of specific and generalized fear memories via dorsal hippocampus CB1 and CB2 receptors. Neuropharmacology 2017, 125, 220–230. [Google Scholar] [CrossRef]
  58. Pertwee, R. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: Δ9-tetrahydrocannabinol, cannabidiol and Δ9-tetrahydrocannabivarin. Br. J. Pharmacol. 2008, 153, 199–215. [Google Scholar] [CrossRef] [Green Version]
  59. Devane, W.A.; Dysarz, F.; Johnson, M.R.; Melvin, L.S.; Howlett, A.C. Determination and characterization of a cannabinoid receptor in rat brain. Mol. Pharmacol. 1988, 34, 605–613. [Google Scholar]
  60. Gururajan, A.; Reif, A.; Cryan, J.F.; Slattery, D.A. The future of rodent models in depression research. Nat. Rev. Neurosci. 2019, 20, 686–701. [Google Scholar] [CrossRef]
  61. Bale, T.L.; Abel, T.; Akil, H.; Carlezon, W.A., Jr.; Moghaddam, B.; Nestler, E.J.; Ressler, K.J.; Thompson, S.M. The critical importance of basic animal research for neuropsychiatric disorders. Neuropsychopharmacology 2019, 44, 1349–1353. [Google Scholar] [CrossRef] [Green Version]
  62. Harro, J. Animal models of depression: Pros and cons. Cell Tissue Res. 2019, 377, 5–20. [Google Scholar] [CrossRef]
  63. Hao, Y.; Ge, H.; Sun, M.; Gao, Y. Selecting an appropriate animal model of depression. Int. J. Mol. Sci. 2019, 20, 4827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wang, Q.; Timberlake, M.A., II; Prall, K.; Dwivedi, Y. The recent progress in animal models of depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2017, 77, 99–109. [Google Scholar] [CrossRef] [PubMed]
  65. Micale, V.; Kucerova, J.; Sulcova, A. Leading compounds for the validation of animal models of psychopathology. Cell Tissue Res. 2013, 354, 309–330. [Google Scholar] [CrossRef] [PubMed]
  66. Willner, P. Validity, reliability and utility of the chronic mild stress model of depression: A 10-year review and evaluation. Psychopharmacology 1997, 134, 319–329. [Google Scholar] [CrossRef]
  67. Slattery, D.A.; Cryan, J.F. Modelling depression in animals: At the interface of reward and stress pathways. Psychopharmacology 2017, 234, 1451–1465. [Google Scholar] [CrossRef]
  68. Bale, T.L.; Epperson, C.N. Sex as a biological variable: Who, what, when, why, and how. Neuropsychopharmacology 2017, 42, 386–396. [Google Scholar] [CrossRef] [Green Version]
  69. Tsou, K.; Brown, S.; Sanudo-Pena, M.C.; Mackie, K.; Walker, J.M. Immunohistochemical distribution of cannabinoid CB1 receptors in the rat central nervous system. Neuroscience 1998, 83, 393–411. [Google Scholar] [CrossRef]
  70. Hu, S.S.; Mackie, K. Distribution of the endocannabinoid system in the central nervous system. Endocannabinoids 2015, 59–93. [Google Scholar]
  71. Zhou, D.; Li, Y.; Tian, T.; Quan, W.; Wang, L.; Shao, Q.; Fu, L.Q.; Zhang, X.H.; Wang, X.Y.; Zhang, H.; et al. Role of the endocannabinoid system in the formation and development of depression. Die Pharm. Int. J. Pharm. Sci. 2017, 72, 435–439. [Google Scholar]
  72. Valverde, O.; Torrens, M. CB1 receptor-deficient mice as a model for depression. Neuroscience 2012, 204, 193–206. [Google Scholar] [CrossRef]
  73. Martin, M.; Ledent, C.; Parmentier, M.; Maldonado, R.; Valverde, O. Involvement of CB1 cannabinoid receptors in emotional behaviour. Psychopharmacology 2002, 159, 379–387. [Google Scholar] [CrossRef] [PubMed]
  74. Shen, C.J.; Zheng, D.; Li, K.X.; Yang, J.M.; Pan, H.Q.; Yu, X.D.; Fu, J.Y.; Zhu, Y.; Sun, Q.X.; Tang, M.Y.; et al. Cannabinoid CB1 receptors in the amygdalar cholecystokinin glutamatergic afferents to nucleus accumbens modulate depressive-like behavior. Nat. Med. 2019, 25, 337–349. [Google Scholar] [CrossRef] [PubMed]
  75. Beyer, C.E.; Dwyer, J.M.; Piesla, M.J.; Platt, B.J.; Shen, R.; Rahman, Z.; Chan, K.; Manners, M.T.; Samad, T.A.; Kennedy, J.D.; et al. Depression-like phenotype following chronic CB1 receptor antagonism. Neurobiol. Dis. 2010, 39, 148–155. [Google Scholar] [CrossRef] [PubMed]
  76. Ebrahimi-Ghiri, M.; Khakpai, F.; Zarrindast, M.R. URB597 abrogates anxiogenic and depressive behaviors in the methamphetamine-withdrawal mice: Role of the cannabinoid receptor type 1, cannabinoid receptor type 2, and transient receptor potential vanilloid 1 channels. J. Psychopharmacol. 2021, 35, 875–884. [Google Scholar] [CrossRef]
  77. McLaughlin, R.J.; Hill, M.N.; Dang, S.S.; Wainwright, S.R.; Galea, L.A.; Hillard, C.J.; Gorzalka, B. Upregulation of CB1 receptor binding in the ventromedial prefrontal cortex promotes proactive stress-coping strategies following chronic stress exposure. Behav. Brain Res. 2013, 237, 333–337. [Google Scholar] [CrossRef] [Green Version]
  78. Rezaie, M.; Nasehi, M.; Vaseghi, S.; Alimohammadzadeh, K.; Vaghar, M.I.; Mohammadi-Mahdiabadi-Hasani, M.H.; Zarrindast, M.R. The interaction effect of sleep deprivation and cannabinoid type 1 receptor in the CA1 hippocampal region on passive avoidance memory, depressive-like behavior and locomotor activity in rats. Behav. Brain Res. 2021, 396, 112901. [Google Scholar] [CrossRef]
  79. Ostadhadi, S.; Haj-Mirzaian, A.; Nikoui, V.; Kordjazy, N.; Dehpour, A.R. Involvement of opioid system in antidepressant-like effect of the cannabinoid CB 1 receptor inverse agonist AM-251 after physical stress in mice. Clin. Exp. Pharmacol. Physiol. 2016, 43, 203–212. [Google Scholar] [CrossRef]
  80. 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]
  81. Maymon, N.; Zer-Aviv, T.M.; Sabban, E.L.; Akirav, I. Neuropeptide Y and cannabinoids interaction in the amygdala after exposure to shock and reminders model of PTSD. Neuropharmacology 2020, 162, 107804. [Google Scholar] [CrossRef]
  82. 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]
  83. de Morais, H.; de Souza, C.P.; da Silva, L.M.; Ferreira, D.M.; Baggio, C.H.; Vanvossen, A.C.; de Carvalho, M.C.; da Silva-Santos, J.E.; Bertoglio, L.J.; Cunha, J.M.; et al. Anandamide reverses depressive-like behavior, neurochemical abnormalities and oxidative-stress parameters in streptozotocin-diabetic rats: Role of CB1 receptors. Eur. Neuropsychopharmacol. 2016, 26, 1590–1600. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, H.N.; Wang, L.; Zhang, R.G.; Chen, Y.C.; Liu, L.; Gao, F.; Nie, H.; Hou, W.G.; Peng, Z.W.; Tan, Q. Anti-depressive mechanism of repetitive transcranial magnetic stimulation in rat: The role of the endocannabinoid system. J. Psychiatr. Res. 2014, 51, 79–87. [Google Scholar] [CrossRef] [PubMed]
  85. Segev, A.; Rubin, A.S.; Abush, H.; Richter-Levin, G.; Akirav, I. Cannabinoid receptor activation prevents the effects of chronic mild stress on emotional learning and LTP in a rat model of depression. Neuropsychopharmacology 2014, 39, 919–933. [Google Scholar] [CrossRef] [PubMed]
  86. Alteba, S.; Zer-Aviv, T.M.; Tenenhaus, A.; David, G.B.; Adelman, J.; Hillard, C.J.; Doron, R.; Akirav, I. Antidepressant-like effects of URB597 and JZL184 in male and female rats exposed to early life stress. Eur. Neuropsychopharmacol. 2020, 39, 70–86. [Google Scholar] [CrossRef] [PubMed]
  87. Chaves, Y.C.; Genaro, K.; Crippa, J.A.; da Cunha, J.M.; Zanoveli, J.M. Cannabidiol induces antidepressant and anxiolytic-like effects in experimental type-1 diabetic animals by multiple sites of action. Metab. Brain Dis. 2021, 36, 639–652. [Google Scholar] [CrossRef] [PubMed]
  88. Hill, M.N.; Gorzalka, B.B. Pharmacological enhancement of cannabinoid CB1 receptor activity elicits an antidepressant-like response in the rat forced swim test. Eur. Neuropsychopharmacol. 2005, 15, 593–599. [Google Scholar] [CrossRef] [PubMed]
  89. 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]
  90. Xu, X.; Wu, K.; Ma, X.; Wang, W.; Wang, H.; Huang, M.; Luo, L.; Su, C.; Yuan, T.; Shi, H.; et al. mGluR5-Mediated eCB Signaling in the Nucleus Accumbens Controls Vulnerability to Depressive-Like Behaviors and Pain after Chronic Social Defeat Stress. Mol. Neurobiol. 2021, 58, 4944–4958. [Google Scholar] [CrossRef]
  91. McLaughlin, R.J.; Hill, M.N.; Bambico, F.R.; Stuhr, K.L.; Gobbi, G.; Hillard, C.J.; Gorzalka, B.B. Prefrontal cortical anandamide signaling coordinates coping responses to stress through a serotonergic pathway. Eur. Neuropsychopharmacol. 2012, 22, 664–671. [Google Scholar] [CrossRef] [Green Version]
  92. Poleszak, E.; Wośko, S.; Sławińska, K.; Wyska, E.; Szopa, A.; Świąder, K.; Wróbel, A.; Doboszewska, U.; Wlaź, P.; Wlaź, A.; et al. Influence of the CB1 and CB2 cannabinoid receptor ligands on the activity of atypical antidepressant drugs in the behavioural tests in mice. Pharmacol. Biochem. Behav. 2020, 188, 172833. [Google Scholar] [CrossRef]
  93. Baur, R.; Gertsch, J.; Sigel, E. The cannabinoid CB1 receptor antagonists rimonabant (SR141716) and AM251 directly potentiate GABAA receptors. Br. J. Pharmacol. 2012, 165, 2479–2484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Rey, A.A.; Purrio, M.; Viveros, M.P.; Lutz, B. Biphasic effects of cannabinoids in anxiety responses: CB1 and GABAB receptors in the balance of GABAergic and glutamatergic neurotransmission. Neuropsychopharmacology 2012, 37, 2624–2634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Calabrese, E.J.; Rubio-Casillas, A. Biphasic effects of THC in memory and cognition. Eur. J. Clin. Investig. 2018, 48, e12920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Sulcova, E.; Mechoulam, R.; Fride, E. Biphasic effects of anandamide. Pharmacol. Biochem. Behav. 1998, 59, 347–352. [Google Scholar] [CrossRef]
  97. Navarrete, F.; García-Gutiérrez, M.S.; Jurado-Barba, R.; Rubio, G.; Gasparyan, A.; Austrich-Olivares, A.; Manzanares, J. Endocannabinoid system components as potential biomarkers in psychiatry. Front. Psychiatry 2020, 315. [Google Scholar] [CrossRef]
  98. Tejeda-Martínez, A.R.; Viveros-Paredes, J.M.; Hidalgo-Franco, G.V.; Pardo-González, E.; Chaparro-Huerta, V.; González-Castañeda, R.E.; Flores-Soto, M.E. Chronic inhibition of FAAH reduces depressive-like behavior and improves dentate gyrus proliferation after chronic unpredictable stress exposure. Behav. Neurol. 2021, 24, 2021. [Google Scholar] [CrossRef]
  99. Realini, N.; Vigano, D.; Guidali, C.; Zamberletti, E.; Rubino, T.; Parolaro, D. Chronic URB597 treatment at adulthood reverted most depressive-like symptoms induced by adolescent exposure to THC in female rats. Neuropharmacology 2011, 60, 235–243. [Google Scholar] [CrossRef]
  100. Bortolato, M.; Mangieri, R.A.; Fu, J.; Kim, J.H.; Arguello, O.; Duranti, A.; Tontini, A.; Mor, M.; Tarzia, G.; Piomelli, D. Antidepressant-like activity of the fatty acid amide hydrolase inhibitor URB597 in a rat model of chronic mild stress. Biol. Psychiatry 2007, 62, 1103–1110. [Google Scholar] [CrossRef] [Green Version]
  101. Jiang, H.X.; Ke, B.W.; Liu, J.; Ma, G.; Hai, K.R.; Gong, D.Y.; Yang, Z.; Zhou, C. Inhibition of fatty acid amide hydrolase improves depressive-like behaviors independent of its peripheral antinociceptive effects in a rat model of neuropathic pain. Anesth. Analg. 2019, 129, 587–597. [Google Scholar] [CrossRef]
  102. Alteba, S.; Korem, N.; Akirav, I. Cannabinoids reverse the effects of early stress on neurocognitive performance in adulthood. Learn. Mem. 2016, 23, 349–358. [Google Scholar] [CrossRef] [Green Version]
  103. Alteba, S.; Portugalov, A.; Hillard, C.J.; Akirav, I. Inhibition of fatty acid amide hydrolase (FAAH) during adolescence and exposure to early life stress may exacerbate depression-like behaviors in male and female rats. Neuroscience 2021, 455, 89–106. [Google Scholar] [CrossRef] [PubMed]
  104. Kruk-Slomka, M.; Michalak, A.; Biala, G. Antidepressant-like effects of the cannabinoid receptor ligands in the forced swimming test in mice: Mechanism of action and possible interactions with cholinergic system. Behav. Brain Res. 2015, 284, 24–36. [Google Scholar] [CrossRef] [PubMed]
  105. Culmer, T.; Dykstra, L. Anandamide (AEA) modifiers indirectly modulate CB1 receptor activity in the forced swim test. FASEB J. 2011, 25, 796. [Google Scholar]
  106. Gobbi, G.; Bambico, F.R.; 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] [Green Version]
  107. Wang, S.; Sun, H.; Liu, S.; Wang, T.; Guan, J.; Jia, J. Role of hypothalamic cannabinoid receptors in post-stroke depression in rats. Brain Res. Bull. 2016, 121, 91–97. [Google Scholar] [CrossRef] [PubMed]
  108. 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]
  109. Sartim, A.G.; Moreira, F.A.; Joca, S.R. Involvement of CB1 and TRPV1 receptors located in the ventral medial prefrontal cortex in the modulation of stress coping behavior. Neuroscience 2017, 340, 126–134. [Google Scholar] [CrossRef]
  110. McLaughlin, R.J.; Hill, M.N.; Morrish, A.C.; Gorzalka, B.B. Local enhancement of cannabinoid CB1 receptor signalling in the dorsal hippocampus elicits an antidepressant-like effect. Behav. Pharmacol. 2007, 18, 431–438. [Google Scholar] [CrossRef]
  111. Piomelli, D. The endocannabinoid system: A drug discovery perspective. Curr. Opin. Investig. Drugs 2005, 6, 672–679. [Google Scholar]
  112. Neumeister, A.; Normandin, M.D.; Pietrzak, R.H.; Piomelli, D.; Zheng, M.Q.; Gujarro-Anton, A.; Potenza, M.N.; Bailey, C.R.; Lin, S.F.; Najafzadeh, S.; et al. Elevated brain cannabinoid CB1 receptor availability in post-traumatic stress disorder: A positron emission tomography study. Mol. Psychiatry 2013, 18, 1034–1040. [Google Scholar] [CrossRef]
  113. Lunn, C.A.; Reich, E.P.; Bober, L. Targeting the CB2 receptor for immune modulation. Expert Opin. Ther. Targets 2006, 10, 653–663. [Google Scholar] [CrossRef] [PubMed]
  114. Kim, J.; Li, Y. Chronic activation of CB2 cannabinoid receptors in the hippocampus increases excitatory synaptic transmission. J. Physiol. 2015, 593, 871–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Ishiguro, H.; Horiuchi, Y.; Tabata, K.; Liu, Q.R.; Arinami, T.; Onaivi, E.S. Cannabinoid CB2 receptor gene and environmental interaction in the development of psychiatric disorders. Molecules 2018, 23, 1836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Quraishi, S.A.; Paladini, C.A. A central move for CB2 receptors. Neuron 2016, 90, 670–671. [Google Scholar] [CrossRef] [Green Version]
  117. Moulton, C.D.; Pickup, J.C.; Ismail, K. The link between depression and diabetes: The search for shared mechanisms. Lancet Diabetes Endocrinol. 2015, 3, 461–471. [Google Scholar] [CrossRef]
  118. Poleszak, E.; Wośko, S.; Sławińska, K.; Wyska, E.; Szopa, A.; Sobczyński, J.; Wróbel, A.; Doboszewska, U.; Wlaź, P.; Wlaź, A.; et al. Ligands of the CB2 cannabinoid receptors augment activity of the conventional antidepressant drugs in the behavioural tests in mice. Behav. Brain Res. 2020, 378, 112297. [Google Scholar] [CrossRef]
  119. Bahi, A.; Al Mansouri, S.; Al Memari, E.; Al Ameri, M.; Nurulain, S.M.; Ojha, S. β-Caryophyllene, a CB2 receptor agonist produces multiple behavioral changes relevant to anxiety and depression in mice. Physiol. Behav. 2014, 135, 119–124. [Google Scholar] [CrossRef]
  120. Hwang, E.S.; Kim, H.B.; Lee, S.; Kim, M.J.; Kim, K.J.; Han, G.; Han, S.Y.; Lee, E.A.; Yoon, J.H.; Kim, D.O.; et al. Antidepressant-like effects of β-caryophyllene on restraint plus stress-induced depression. Behav. Brain Res. 2020, 380, 112439. [Google Scholar] [CrossRef]
  121. Aguilar-Ávila, D.S.; Flores-Soto, M.E.; Tapia-Vázquez, C.; Pastor-Zarandona, O.A.; López-Roa, R.I.; Viveros-Paredes, J.M. β-Caryophyllene, a natural sesquiterpene, attenuates neuropathic pain and depressive-like behavior in experimental diabetic mice. J. Med. Food 2019, 22, 460–468. [Google Scholar] [CrossRef]
  122. Hu, B.; Doods, H.; Treede, R.D.; Ceci, A. Depression-like behaviour in rats with mononeuropathy is reduced by the CB2-selective agonist GW405833. PAIN 2009, 143, 206–212. [Google Scholar] [CrossRef]
  123. García-Gutiérrez, M.S.; Pérez-Ortiz, J.M.; Gutiérrez-Adán, A.; Manzanares, J. Depression-resistant endophenotype in mice overexpressing cannabinoid CB2 receptors. Br. J. Pharmacol. 2010, 160, 1773–1784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Lee, D.K.; Lynch, K.R.; Nguyen, T.; Im, D.S.; Cheng, R.; Saldivia, V.R.; Liu, Y.; Liu, I.S.; Heng, H.H.; Seeman, P.; et al. Cloning and characterization of additional members of the G protein-coupled receptor family. Biochim. Et Biophys. Acta (BBA)-Gene Struct. Expr. 2000, 1490, 311–323. [Google Scholar] [CrossRef]
  125. Wróbel, A.; Serefko, A.; Szopa, A.; Ulrich, D.; Poleszak, E.; Rechberger, T. O-1602, an agonist of atypical Cannabinoid receptors GPR55, reverses the symptoms of depression and detrusor overactivity in rats subjected to corticosterone treatment. Front. Pharmacol. 2020, 11, 1002. [Google Scholar] [CrossRef] [PubMed]
  126. Shen, S.; Yu, R.; Li, W.; Liang, L.F.; Han, Q.; Huang, H.; Li, B.; Xu, S.; Wu, G.; Zhang, Y.Q.; et al. The Protective Effects of GPR55 against Hippocampal Neuroinflammation and Neurogenic Damage in CSDS Mice; Research Square: Durham, NC, USA, 2021. [Google Scholar]
  127. Huang, W.; Ke, Y.; Chen, R. Region-specific dysregulation of endocannabinoid system in learned helplessness model of depression. Neuroreport 2021, 32, 345–351. [Google Scholar] [CrossRef] [PubMed]
  128. Trevisani, M.; Szallasi, A. Targeting TRPV1: Challenges and issues in pain management. Open Drug Discov. J. 2014, 2, 37–49. [Google Scholar] [CrossRef]
  129. Li, J.X. Pain and depression comorbidity: A preclinical perspective. Behav. Brain Res. 2015, 276, 92–98. [Google Scholar] [CrossRef] [Green Version]
  130. Abdelhamid, R.E.; Kovács, K.J.; Nunez, M.G.; Larson, A.A. Depressive behavior in the forced swim test can be induced by TRPV1 receptor activity and is dependent on NMDA receptors. Pharmacol. Res. 2014, 79, 21–27. [Google Scholar] [CrossRef] [Green Version]
  131. Navarria, A.; Tamburella, A.; Iannotti, F.A.; Micale, V.; Camillieri, G.; Gozzo, L.; Verde, R.; Imperatore, R.; Leggio, G.M.; Drago, F.; et al. The dual blocker of FAAH/TRPV1 N-arachidonoylserotonin reverses the behavioral despair induced by stress in rats and modulates the HPA-axis. Pharmacol. Res. 2014, 87, 151–159. [Google Scholar] [CrossRef]
  132. Kirkedal, C.; Wegener, G.; Moreira, F.; Joca, S.R.; Liebenberg, N. A dual inhibitor of FAAH and TRPV1 channels shows dose-dependent effect on depression-like behaviour in rats. Acta Neuropsychiatr. 2017, 29, 324–329. [Google Scholar] [CrossRef]
  133. 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] [Green Version]
  134. Abame, M.A.; He, Y.; Wu, S.; Xie, Z.; Zhang, J.; Gong, X.; Wu, C.; Shen, J. Chronic administration of synthetic cannabidiol induces antidepressant effects involving modulation of serotonin and noradrenaline levels in the hippocampus. Neurosci. Lett. 2021, 744, 135594. [Google Scholar] [CrossRef] [PubMed]
  135. Bis-Humbert, C.; García-Cabrerizo, R.; García-Fuster, M.J. Decreased sensitivity in adolescent versus adult rats to the antidepressant-like effects of cannabidiol. Psychopharmacology 2020, 237, 1621–1631. [Google Scholar] [CrossRef] [PubMed]
  136. de Morais, H.; Chaves, Y.C.; Waltrick, A.P.; Jesus, C.H.; Genaro, K.; Crippa, J.A.; da Cunha, J.M.; Zanoveli, J.M. Sub-chronic treatment with cannabidiol but not with URB597 induced a mild antidepressant-like effect in diabetic rats. Neurosci. Lett. 2018, 682, 62–68. [Google Scholar] [CrossRef] [PubMed]
  137. Gáll, Z.; Farkas, S.; Albert, Á.; Ferencz, E.; Vancea, S.; Urkon, M.; Kolcsár, M. Effects of chronic cannabidiol treatment in the rat chronic unpredictable mild stress model of depression. Biomolecules 2020, 10, 801. [Google Scholar] [CrossRef]
  138. Shbiro, L.; Hen-Shoval, D.; Hazut, N.; Rapps, K.; Dar, S.; Zalsman, G.; Mechoulam, R.; Weller, A.; Shoval, G. Effects of cannabidiol in males and females in two different rat models of depression. Physiol. Behav. 2019, 201, 59–63. [Google Scholar] [CrossRef]
  139. Sales, A.J.; Crestani, C.C.; Guimarães, F.S.; Joca, S.R. Antidepressant-like effect induced by Cannabidiol is dependent on brain serotonin levels. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 86, 255–261. [Google Scholar] [CrossRef] [Green Version]
  140. Sales, A.J.; Fogaça, M.V.; Sartim, A.G.; Pereira, V.S.; Wegener, G.; Guimarães, F.S.; Joca, S.R. Cannabidiol induces rapid and sustained antidepressant-like effects through increased BDNF signaling and synaptogenesis in the prefrontal cortex. Mol. Neurobiol. 2019, 56, 1070–1081. [Google Scholar] [CrossRef]
  141. Sales, A.J.; Guimarães, F.S.; Joca, S.R. CBD modulates DNA methylation in the prefrontal cortex and hippocampus of mice exposed to forced swim. Behav. Brain Res. 2020, 388, 112627. [Google Scholar] [CrossRef]
  142. Réus, G.Z.; Stringari, R.B.; Ribeiro, K.F.; Luft, T.; Abelaira, H.M.; Fries, G.R.; Aguiar, B.W.; Kapczinski, F.; Hallak, J.E.; Zuardi, A.W.; et al. Administration of cannabidiol and imipramine induces antidepressant-like effects in the forced swimming test and increases brain-derived neurotrophic factor levels in the rat amygdala. Acta Neuropsychiatr. 2011, 23, 241–248. [Google Scholar] [CrossRef]
  143. Xu, C.; Chang, T.; Du, Y.; Yu, C.; Tan, X.; Li, X. Pharmacokinetics of oral and intravenous cannabidiol and its antidepressant-like effects in chronic mild stress mouse model. Environ. Toxicol. Pharmacol. 2019, 70, 103202. [Google Scholar] [CrossRef]
  144. Malvestio, R.B.; Medeiros, P.; Negrini-Ferrari, S.E.; Oliveira-Silva, M.; Medeiros, A.C.; Padovan, C.M.; Luongo, L.; Maione, S.; Coimbra, N.C.; de Freitas, R.L. Cannabidiol in the prelimbic cortex modulates the comorbid condition between the chronic neuropathic pain and depression-like behaviour in rats: The role of medial prefrontal cortex 5-HT1A and CB1 receptors. Brain Res. Bull. 2021, 174, 323–338. [Google Scholar] [CrossRef] [PubMed]
  145. Zanelati, T.V.; Biojone, C.; Moreira, F.A.; Guimarães, F.S.; Joca, S.R. Antidepressant-like effects of cannabidiol in mice: Possible involvement of 5-HT1A receptors. Br. J. Pharmacol. 2010, 159, 122–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Linge, R.; Jiménez-Sánchez, L.; Campa, L.; Pilar-Cuéllar, F.; Vidal, R.; Pazos, A.; Adell, A.; Díaz, Á. Cannabidiol induces rapid-acting antidepressant-like effects and enhances cortical 5-HT/glutamate neurotransmission: Role of 5-HT1A receptors. Neuropharmacology 2016, 103, 16–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Sartim, A.G.; Guimarães, F.S.; Joca, S.R. Antidepressant-like effect of cannabidiol injection into the ventral medial prefrontal cortex—Possible involvement of 5-HT1A and CB1 receptors. Behav. Brain Res. 2016, 303, 218–227. [Google Scholar] [CrossRef]
  148. Fogaça, M.V.; Campos, A.C.; Coelho, L.D.; Duman, R.S.; Guimarães, F.S. The anxiolytic effects of cannabidiol in chronically stressed mice are mediated by the endocannabinoid system: Role of neurogenesis and dendritic remodeling. Neuropharmacology 2018, 135, 22–33. [Google Scholar] [CrossRef]
  149. 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]
  150. 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. [Google Scholar] [CrossRef]
  151. McLaughlin, P.J. Reports of the death of CB1 antagonists have been greatly exaggerated: Recent preclinical findings predict improved safety in the treatment of obesity. Behav. Pharmacol. 2012, 23, 537–550. [Google Scholar] [CrossRef]
  152. Kruger, J.S.; Blavos, A.; Castor, T.S.; Wotring, A.J.; Wagner-Greene, V.R.; Glassman, T.; Kruger, D.J. Manipulation checking the munchies: Validating self-reported dietary behaviors during cannabis intoxication. Hum. Ethol. 2019, 34, 10–16. [Google Scholar] [CrossRef]
  153. 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]
  154. Soyka, M. Rimonabant and depression. Pharmacopsychiatry 2008, 41, 204–205. [Google Scholar] [CrossRef] [PubMed]
  155. Martin, A.; Naunton, M.; Kosari, S.; Peterson, G.; Thomas, J.; Christenson, J.K. Treatment guidelines for PTSD: A systematic review. J. Clin. Med. 2021, 10, 4175. [Google Scholar] [CrossRef] [PubMed]
  156. Aragona, M.; Onesti, E.; Tomassini, V.; Conte, A.; Gupta, S.; Gilio, F.; Pantano, P.; Pozzilli, C.; Inghilleri, M. Psychopathological and cognitive effects of therapeutic cannabinoids in multiple sclerosis: A double-blind, placebo controlled, crossover study. Clin. Neuropharmacol. 2009, 32, 41–47. [Google Scholar] [CrossRef] [PubMed]
  157. Alessandria, G.; Meli, R.; Infante, M.T.; Vestito, L.; Capello, E.; Bandini, F. Long-term assessment of the cognitive effects of nabiximols in patients with multiple sclerosis: A pilot study. Clin. Neurol. Neurosurg. 2020, 196, 105990. [Google Scholar] [CrossRef]
  158. Portenoy, R.K.; Ganae-Motan, E.D.; Allende, S.; Yanagihara, R.; Shaiova, L.; Weinstein, S.; McQuade, R.; Wright, S.; Fallon, M.T. Nabiximols for opioid-treated cancer patients with poorly-controlled chronic pain: A randomized, placebo-controlled, graded-dose trial. J. Pain 2012, 13, 438–449. [Google Scholar] [CrossRef]
  159. 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] [Green Version]
  160. 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]
  161. Frank, B.; Serpell, M.G.; Hughes, J.; Matthews, J.N.; Kapur, D. Comparison of analgesic effects and patient tolerability of nabilone and dihydrocodeine for chronic neuropathic pain: Randomised, crossover, double blind study. BMJ 2008, 336, 199–201. [Google Scholar] [CrossRef] [Green Version]
  162. Narang, S.; Gibson, D.; Wasan, A.D.; Ross, E.L.; Michna, E.; Nedeljkovic, S.S.; Jamison, R.N. Efficacy of dronabinol as an adjuvant treatment for chronic pain patients on opioid therapy. J. Pain 2008, 9, 254–264. [Google Scholar] [CrossRef]
  163. Deckman, T.; DeWall, C.N.; Way, B.; Gilman, R.; Richman, S. Can marijuana reduce social pain? Soc. Psychol. Personal. Sci. 2014, 5, 131–139. [Google Scholar] [CrossRef]
  164. Solowij, N.; Broyd, S.J.; Beale, C.; Prick, J.A.; Greenwood, L.M.; Van Hell, H.; Suo, C.; Galettis, P.; Pai, N.; Fu, S.; et al. Therapeutic effects of prolonged cannabidiol treatment on psychological symptoms and cognitive function in regular cannabis users: A pragmatic open-label clinical trial. Cannabis Cannabinoid Res. 2018, 3, 21–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Allsop, D.J.; Copeland, J.; Lintzeris, N.; Dunlop, A.J.; Montebello, M.; Sadler, C.; Rivas, G.R.; Holland, R.M.; Muhleisen, P.; Norberg, M.M.; et al. Nabiximols as an agonist replacement therapy during cannabis withdrawal: A randomized clinical trial. JAMA Psychiatry 2014, 71, 281–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Babson, K.A.; Boden, M.T.; Bonn-Miller, M.O. Sleep quality moderates the relation between depression symptoms and problematic cannabis use among medical cannabis users. Am. J. Drug Alcohol Abus. 2013, 39, 211–216. [Google Scholar] [CrossRef] [PubMed]
  167. Denson, T.F.; Earleywine, M. Decreased depression in marijuana users. Addict. Behav. 2006, 31, 738–742. [Google Scholar] [CrossRef] [PubMed]
  168. Walsh, Z.; Gonzalez, R.; Crosby, K.; Thiessen, M.S.; Carroll, C.; Bonn-Miller, M.O. Medical cannabis and mental health: A guided systematic review. Clin. Psychol. Rev. 2017, 51, 15–29. [Google Scholar] [CrossRef] [PubMed]
  169. Bahorik, A.L.; Sterling, S.A.; Campbell, C.I.; Weisner, C.; Ramo, D.; Satre, D.D. Medical and non-medical marijuana use in depression: Longitudinal associations with suicidal ideation, everyday functioning, and psychiatry service utilization. J. Affect. Disord. 2018, 241, 8–14. [Google Scholar] [CrossRef] [PubMed]
  170. Schlienz, N.J.; Scalsky, R.; Martin, E.L.; Jackson, H.; Munson, J.; Strickland, J.C.; Bonn-Miller, M.O.; Loflin, M.; Vandrey, R. A cross-sectional and prospective comparison of medicinal cannabis users and controls on self-reported health. Cannabis Cannabinoid Res. 2021, 6, 548–558. [Google Scholar] [CrossRef]
  171. Rana, T.; Behl, T.; Sehgal, A.; Mehta, V.; Singh, S.; Kumar, R.; Bungau, S. Integrating endocannabinoid signalling in depression. J. Mol. Neurosci. 2021, 71, 2022–2034. [Google Scholar] [CrossRef]
  172. Hill, M.N.; Miller, G.E.; Carrier, E.J.; Gorzalka, B.B.; Hillard, C.J. Circulating endocannabinoids and N-acyl ethanolamines are differentially regulated in major depression and following exposure to social stress. Psychoneuroendocrinology 2009, 34, 1257–1262. [Google Scholar] [CrossRef] [Green Version]
  173. Hill, M.N.; Miller, G.E.; Ho, W.S.; Gorzalka, B.B.; Hillard, C.J. Serum endocannabinoid content is altered in females with depressive disorders: A preliminary report. Pharmacopsychiatry 2008, 41, 48–53. [Google Scholar] [CrossRef] [Green Version]
  174. Romero-Sanchiz, P.; Nogueira-Arjona, R.; Pastor, A.; Araos, P.; Serrano, A.; Boronat, A.; Garcia-Marchena, N.; Mayoral, F.; Bordallo, A.; Alen, F.; et al. Plasma concentrations of oleoylethanolamide in a primary care sample of depressed patients are increased in those treated with selective serotonin reuptake inhibitor-type antidepressants. Neuropharmacology 2019, 149, 212–220. [Google Scholar] [CrossRef] [PubMed]
  175. Heyman, E.; Gamelin, F.X.; Goekint, M.; Piscitelli, F.; Roelands, B.; Leclair, E.; Di Marzo, V.; Meeusen, R. Intense exercise increases circulating endocannabinoid and BDNF levels in humans—possible implications for reward and depression. Psychoneuroendocrinology 2012, 37, 844–851. [Google Scholar] [CrossRef] [PubMed]
  176. Meyer, J.D.; Crombie, K.M.; Cook, D.B.; Hillard, C.J.; Koltyn, K.F. Serum endocannabinoid and mood changes after exercise in major depressive disorder. Med. Sci. Sports Exerc. 2019, 51, 1909. [Google Scholar] [CrossRef] [Green Version]
  177. Kranaster, L.; Hoyer, C.; Aksay, S.S.; Bumb, J.M.; Leweke, F.M.; Ke, C.; Thiel, M.; Lutz, B.; Bindila, L.; Sartorius, A. Electroconvulsive therapy enhances endocannabinoids in the cerebrospinal fluid of patients with major depression: A preliminary prospective study. Eur. Arch. Psychiatry Clin. Neurosci. 2017, 267, 781–786. [Google Scholar] [CrossRef] [PubMed]
  178. Choi, K.; Le, T.; McGuire, J.; Xing, G.; Zhang, L.; Li, H.; Parker, C.C.; Johnson, L.R.; Ursano, R.J. Expression pattern of the cannabinoid receptor genes in the frontal cortex of mood disorder patients and mice selectively bred for high and low fear. J. Psychiatr. Res. 2012, 46, 882–889. [Google Scholar] [CrossRef] [PubMed]
  179. Hungund, B.L.; Vinod, K.Y.; Kassir, S.A.; Basavarajappa, B.S.; Yalamanchili, R.; Cooper, T.B.; Mann, J.J.; Arango, V. Upregulation of CB1 receptors and agonist-stimulated [35S] GTPγS binding in the prefrontal cortex of depressed suicide victims. Mol. Psychiatry 2004, 9, 184–190. [Google Scholar] [CrossRef]
  180. 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. 2007, 114, 1055–1063. [Google Scholar] [CrossRef]
  181. Leweke, F.M.; Koethe, D. Cannabis and psychiatric disorders: It is not only addiction. Addict. Biol. 2008, 13, 264–275. [Google Scholar] [CrossRef]
  182. Hirvonen, J.; Goodwin, R.S.; Li, C.T.; Terry, G.E.; Zoghbi, S.S.; Morse, C.; Pike, V.W.; Volkow, N.D.; Huestis, M.A.; Innis, R. Reversible and regionally selective downregulation of brain cannabinoid CB1 receptors in chronic daily cannabis smokers. Mol. Psychiatry 2012, 17, 642–649. [Google Scholar] [CrossRef] [Green Version]
  183. Klugmann, M.; Klippenstein, V.; Leweke, F.M.; Spanagel, R.; Schneider, M. Cannabinoid exposure in pubertal rats increases spontaneous ethanol consumption and NMDA receptor associated protein levels. Int. J. Neuropsychopharmacol. 2011, 14, 505–517. [Google Scholar] [CrossRef] [Green Version]
  184. Kong, X.; Miao, Q.; Lu, X.; Zhang, Z.; Chen, M.; Zhang, J.; Zhai, J. The association of endocannabinoid receptor genes (CNR1 and CNR2) polymorphisms with depression: A meta-analysis. Medicine 2019, 98. [Google Scholar] [CrossRef] [PubMed]
  185. Yao, Y.; Xu, Y.; Zhao, J.; Ma, Y.; Su, K.; Yuan, W.; Ma, J.Z.; Payne, T.J.; Li, M.D. Detection of significant association between variants in cannabinoid receptor 1 Gene (CNR1) and personality in African–American population. Front. Genet. 2018, 9, 199. [Google Scholar] [CrossRef] [PubMed]
  186. Icick, R.; Peoc’h, K.; Karsinti, E.; Ksouda, K.; Hajj, A.; Bloch, V.; Prince, N.; Mouly, S.; Bellivier, F.; Lépine, J.P.; et al. A cannabinoid receptor 1 polymorphism is protective against major depressive disorder in methadone-maintained outpatients. Am. J. Addict. 2015, 24, 613–620. [Google Scholar] [CrossRef]
  187. Tsai, S.J.; Wang, Y.C.; Hong, C.J. Association study between cannabinoid receptor gene (CNR1) and pathogenesis and psychotic symptoms of mood disorders. Am. J. Med. Genet. 2001, 105, 219–221. [Google Scholar] [CrossRef] [PubMed]
  188. Mitjans, M.; Serretti, A.; Fabbri, C.; Gastó, C.; Catalán, R.; Fañanás, L.; Arias, B. Screening genetic variability at the CNR1 gene in both major depression etiology and clinical response to citalopram treatment. Psychopharmacology 2013, 227, 509–519. [Google Scholar] [CrossRef]
  189. Monteleone, P.; Bifulco, M.; Maina, G.; Tortorella, A.; Gazzerro, P.; Proto, M.C.; Di Filippo, C.; Monteleone, F.; Canestrelli, B.; Buonerba, G.; et al. Investigation of CNR1 and FAAH endocannabinoid gene polymorphisms in bipolar disorder and major depression. Pharmacol. Res. 2010, 61, 400–404. [Google Scholar] [CrossRef] [PubMed]
  190. Lazary, J.; Eszlari, N.; Juhasz, G.; Bagdy, G. A functional variant of CB2 receptor gene interacts with childhood trauma and FAAH gene on anxious and depressive phenotypes. J. Affect. Disord. 2019, 257, 716–722. [Google Scholar] [CrossRef] [PubMed]
  191. Onaivi, E.S.; Ishiguro, H.; Gong, J.P.; Patel, S.; Meozzi, P.A.; Myers, L.; Perchuk, A.; Mora, Z.; Tagliaferro, P.A.; Gardner, E.; et al. Brain neuronal CB2 cannabinoid receptors in drug abuse and depression: From mice to human subjects. PLoS ONE 2008, 3, e1640. [Google Scholar] [CrossRef] [Green Version]
  192. Ishiguro, H.; Iwasaki, S.; Teasenfitz, L.; Higuchi, S.; Horiuchi, Y.; Saito, T.; Arinami, T.; Onaivi, E.S. Involvement of cannabinoid CB2 receptor in alcohol preference in mice and alcoholism in humans. Pharm. J. 2007, 7, 380–385. [Google Scholar] [CrossRef] [Green Version]
  193. Petrakis, I.L.; Gonzalez, G.; Rosenheck, R.; Krystal, J.H. Comorbidity of alcoholism and psychiatric disorders: An overview. Alcohol Res. Health 2002, 26, 81. [Google Scholar]
  194. Minocci, D.A.; Massei, J.; Martino, A.; Milianti, M.; Piz, L.; Di Bello, D.; Sbrana, A.; Martinotti, E.; Rossi, A.M.; Nieri, P. Genetic association between bipolar disorder and 524A > C (Leu133Ile) polymorphism of CNR2 gene, encoding for CB2 cannabinoid receptor. J. Affect. Disord. 2011, 134, 427–430. [Google Scholar] [CrossRef] [PubMed]
  195. García-Gutiérrez, M.S.; Navarrete, F.; Navarro, G.; Reyes-Resina, I.; Franco, R.; Lanciego, J.L.; Giner, S.; Manzanares, J. Alterations in gene and protein expression of cannabinoid CB2 and GPR55 receptors in the dorsolateral prefrontal cortex of suicide victims. Neurotherapeutics 2018, 15, 796–806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Lazary, J.; Eszlari, N.; Juhasz, G.; Bagdy, G. Genetically reduced FAAH activity may be a risk for the development of anxiety and depression in persons with repetitive childhood trauma. Eur. Neuropsychopharmacol. 2016, 26, 1020–1028. [Google Scholar] [CrossRef] [PubMed]
  197. Maple, K.E.; McDaniel, K.A.; Shollenbarger, S.G.; Lisdahl, K.M. Dose-dependent cannabis use, depressive symptoms, and FAAH genotype predict sleep quality in emerging adults: A pilot study. Am. J. Drug Alcohol Abus. 2016, 42, 431–440. [Google Scholar] [CrossRef] [Green Version]
  198. Turecki, G. Dissecting the suicide phenotype: The role of impulsive–aggressive behaviours: 2003 CCNP Young Investigator Award Paper. J. Psychiatry Neurosci. 2005, 30, 398–408. [Google Scholar]
  199. Mammen, G.; Rueda, S.; Roerecke, M.; Bonato, S.; Lev-Ran, S.; Rehm, J. Association of cannabis with long-term clinical symptoms in anxiety and mood disorders: A systematic review of prospective studies. J. Clin. Psychiatry 2018, 79, 2248. [Google Scholar] [CrossRef]
  200. Cuttler, C.; Spradlin, A.; McLaughlin, R.J. A naturalistic examination of the perceived effects of cannabis on negative affect. J. Affect. Disord. 2018, 235, 198–205. [Google Scholar] [CrossRef]
  201. Henquet, C.; Rosa, A.; Krabbendam, L.; Papiol, S.; Faňanás, L.; Drukker, M.; Ramaekers, J.G.; van Os, J. An experimental study of catechol-O-methyltransferase Val158Met moderation of Δ-9-tetrahydrocannabinol-induced effects on psychosis and cognition. Neuropsychopharmacology 2006, 31, 2748–2757. [Google Scholar] [CrossRef]
  202. World Health Organization: The Health and Social Effects of Nonmedical Cannabis Use: World Health Organization. Available online: https://apps.who.int/iris/handle/10665/251056 (accessed on 29 March 2022).
Table 1. A summary of the findings regarding the effects of CB1 antagonists on depression-like behavior in rodents.
Table 1. A summary of the findings regarding the effects of CB1 antagonists on depression-like behavior in rodents.
DrugAdministrationAnimalsStressModelEffectReference
AM251
(CB1 antagonist)
Acute,
1 μg,
i.c.v.
Male NMRI mice-FSTElevated immobility[76]
Acute,
0.28 ng,
PFC microinjection
Male SD ratsCUSFSTElevated immobility[77]
Acute,
0.01 ng,
HIPP microinjection
Male Wistar ratsSleep deprivationFSTElevated immobility[78]
Acute,
0.3, 0.5 mg/kg,
i.p.
Male NMRI miceFoot shockFSTDecreased immobility[79]
Acute,
0.3, 0.5 mg/kg,
i.p.
Male NMRI miceFoot shockTSTDecreased immobility[79]
Acute,
0.3, 0.5, 1, 10 mg/kg,
i.p.
C57BL/6 male mice-FSTDecreased immobility[80]
Acute,
0.3, 0.5, 1 mg/kg,
i.p.
C57BL/6 male mice-TSTDecreased immobility[80]
Acute,
0.01 μg,
BLA microinjection
Male SD rats-FSTDecreased immobility[81]
Rimonabant
(CB1 antagonist)
Chronic (21 days),
10 mg/kg,
i.p.
Male SD rats-FSTElevated immobility[75]
-SPTDecreased sucrose preference[75]
Acute (2 times),
3 mg/kg, 10 mg/kg,
oral
Male SD and Wistar rats-FSTDecreased immobility[82]
Chronic (35 days),
10 mg/kg,
oral
OF1 miceCMSFSTDecreased immobility[82]
rTMS: repetitive transcranial magnetic stimulation; CUMS: chronic unpredictable mild stress; CMS: chronic mild stress; ELS: early life stress; CHPG: (RS)-2-chloro-5-hydroxyphenylglycine; TST: Tail suspension test; i.c.v.: intracerebroventricular; CMS: chronic mild stress; HIPP: hippocampus; NMRI: Naval Medical Research Institute; CCI: chronic constriction injury; NP: neuropathic pain; SD: Sprague–Dawley; SPT: sucrose preference test; SaPT: saccharine preference test.
Table 2. A summary of the effects of CB1 antagonists co-administered with cannabinoid agonists on depression-like behavior in rodents.
Table 2. A summary of the effects of CB1 antagonists co-administered with cannabinoid agonists on depression-like behavior in rodents.
DrugAdministrationAnimalsStressTreatmentModelEffectReference
AM251
(CB1 antagonist)
Acute,
1 mg/kg,
i.p.
Male Wistar rats Streptozotocin (diabetic)AEAFSTElevated immobility[83]
7 days,
1 mg/kg,
i.p.
Male SD rat CUMSrTMSFSTElevated immobility[84]
3 days,
0.3 mg/kg,
i.p.
Male SD ratsCMSWIN55,212-2FSTElevated immobility[85]
14 days,
0.3 mg/kg,
i.p.
Acute,
1 mg/kg,
i.p.
Male and female SD ratsELSJZL184 or URB597FSTElevated immobility[86]
Male Wistar ratsStreptozotocin (diabetic)CBDFSTElevated immobility[87]
Acute,
5 mg/kg,
i.p.
Male Long-Evans rats-AM404FSTElevated immobility[88]
Acute,
0.8 μg,
NAc microinjection
Male C57BL/6J miceSocial defeatCHPGTSTElevated immobility[90]
Acute,
0.28 ng,
PFC microinjection
Male SD rats-URB597FSTElevated immobility[91]
Acute,
0.25 mg/kg,
i.p
Male Albino Swiss mice-TianeptineFSTDecreasedimmobility[92]
Rimonabant
(CB1 antagonist)
Acute,
3 mg/kg,
i.p.
Male Wistar rats-URB597, AM404, CP55,940FSTElevated immobility[89]
rTMS: repetitive transcranial magnetic stimulation; CUMS: chronic unpredictable mild stress; CMS: chronic mild stress; ELS: early life stress; CHPG: (RS)-2-chloro-5-hydroxyphenylglycine; TST: Tail suspension test; i.c.v..: intracerebroventricular; CMS: chronic mild stress; HIPP: hippocampus; NMRI: Naval Medical Research Institute; CCI: chronic constriction injury; NP: neuropathic pain; SPT: sucrose preference test; SaPT: saccharine preference test; SD: Sprague–Dawley.
Table 3. A summary of CB1-mediated effects of cannabinoids on depression-like behavior in rodents.
Table 3. A summary of CB1-mediated effects of cannabinoids on depression-like behavior in rodents.
DrugAdministrationAnimalsStressModelEffectReference
URB597
(FAAH Inhibitor
Chronic,
0.2 mg/kg,
i.p.
C57BL/6J miceCUSFSTDecreased immobility[98]
Chronic,
0.3 mg/kg,
i.p.
Female SD ratsAdolescent THCFSTDecreased immobility[99]
Chronic,
0.3 mg/kg,
i.p.
Female SD ratsAdolescent THCSPTElevated sucrose preference[99]
Chronic,
0.3 mg/kg,
i.p.
Male Wistar ratsCMSSPTElevated sucrose preference[100]
Chronic,
5.8 mg/kg,
i.p.
14 days (during mid-adolescence),
0.4 mg/kg,
i.p.
Male Wistar ratsCCI injury (NP)FSTDecreased immobility[101]
Male and female SD ratsELSFSTDecreased immobility[86]
14 days (during late-adolescence),
0.4 mg/kg,
i.p.
Male and female SD ratsELSFSTElevated immobility[103]
Acute,
0.3 mg/kg,
i.p.
Male SD rats Severe shockFSTDecreased immobility[40]
Acute,
0.3 mg/kg,
i.p.
Male SD ratsSevere shockSaPTElevated saccharine preference[40]
Acute,
0.03, 0.1, 0.3 mg/kg,
i.p.
Male Wistar rats-FSTDecreased immobility[89]
Acute,
0.1 mg/kg,
i.p.
Male C57BL/6 mice-FSTDecreased immobility[106]
Acute,
0.1 mg/kg,
i.p.
Male C57BL/6 mice-TSTDecreased immobility[106]
Acute,
1, 3.2 mg/kg,
i.p.
Male SD rats-FSTDecreased immobility[105]
Acute,
5, 10 ng,
i.c.v.
NMRI miceMethamphetamineFSTDecreased immobility[76]
Acute,
0.05, 0.1, 1, 5, 10 μg,
i.c.v.
Male Swiss mice-FSTDecreased immobility[108]
Acute,
0.01, 0.1, 1 nmol,
vmPFC microinjection
Male Wistar rats-FSTDecreased immobility[109]
Acute,
0.01 μg,
PFC microinjection
Male Wistar rats-FSTDecreased immobility[91]
HU-210
(CB1/CB2 agonist)
Acute,
0.5, 1 μg,
dentate gyrus microinjection
Male Wistar rats-FSTDecreased immobility[110]
AM404
(AEA reuptake inhibitor)
Acute,
5 mg/kg,
i.p.
Male Long-Evans rats
Rats
-FSTDecreased immobility[88]
Acute,
0.1, 0.3, 1, 3 mg/kg,
i.p.
-FSTDecreased immobility[89]
Acute,
1 mg/kg,
i.p.
Male SD rats-FSTDecreased immobility[105]
Acute,
0.1, 1, 5, 10 μg,
i.c.v.
Male Swiss mice-FSTDecreased immobility[108]
CP55,940
(CB1/CB2 agonist)
Acute,
0.03, 0.1, 0.3 mg/kg,
i.p.
Male Wistar rats-FSTDecreased immobility[89]
OleamideAcute,
10, 20 mg/kg,
i.p.
Male Swiss mice-FSTDecreased immobility[104]
rTMS: repetitive transcranial magnetic stimulation; CUMS: chronic unpredictable mild stress; CMS: chronic mild stress; ELS: early life stress; CHPG: (RS)-2-chloro-5-hydroxyphenylglycine; TST: Tail suspension test; i.c.v.: intracerebroventricular; CMS: chronic mild stress; HIPP: hippocampus; NMRI: Naval Medical Research Institute; CCI: chronic constriction injury; NP: neuropathic pain; SPT: sucrose preference test; SaPT: saccharine preference test; SD: Sprague–Dawley; ACEA: arachidonyl-2-chloroethylamide.
Table 4. A summary of the effects of CBD on depression-like behavior in rodents.
Table 4. A summary of the effects of CBD on depression-like behavior in rodents.
CBD AdministrationAnimalsStressModelEffectReference
Acute,
200 mg/kg,
i.p.
Male Swiss Webster mice-FSTDecreased immobility[133]
7-day,
100 mg/kg,
i.p.
Male C57BL/6J mice-FSTDecreased immobility[134]
7-day,
10, 30 mg/kg,
i.p.
Male SD rats-FSTDecreased immobility[135]
Sub-chronic,
30 mg/kg,
i.p.
Male Wistar ratsStreptozotocin (diabetic)FSTDecreased immobility[136]
Chronic,
10 mg/kg,
i.p.
Male Wistar ratsCUMSSPTElevated sucrose preference[137]
Acute,
30 mg/kg,
oral
Male and female WKY ratsWKY (genetic model)SaPTElevated saccharine preference[138]
Acute,
30 mg/kg,
oral
Male and female WKY and male FSL ratsWKY or FSL (genetic models)FSTDecreased immobility[138]
Acute,
30 mg/kg,
oral
Male WKY ratsWKY (genetic model)SaPTElevated saccharine preference[39]
Acute,
10 mg/kg,
i.p.
Male Swiss mice-FSTDecreased immobility[139]
Acute,
7 mg/kg,
i.p.
(co-administered with fluoxetine)
Male Swiss mice-FSTDecreased immobility[139]
Acute,
10 mg/kg,
i.p.
Male Swiss mice-FSTDecreased immobility[141]
Acute,
7 mg/kg,
i.p.
(co-administered with AzaD or RG108)
Male Swiss mice-FSTDecreased immobility[141]
Acute,
10 mg/kg,
i.p.
Male Swiss mice-FSTDecreased immobility[140]
Chronic,
30 mg/kg,
i.p.
Male Wistar rats-FSTDecreased immobility[142]
Acute,
30 mg/kg,
i.p.
Male Wistar rats-FSTDecreased immobility[142]
Acute,
10 mg/kg,
i.v.
Male ICR miceCMSFSTDecreased immobility[143]
Acute,
100 mg/kg,
oral
Male ICR miceCMSFSTDecreased immobility[143]
Acute,
15, 30, 60 nmol,
mPFC microinjection
Male Wistar ratsCCI injury (NP)FSTDecreased immobility[144]
Acute,
30 mg/kg,
i.p.
Male Swiss mice-FSTDecreased immobility[145]
7 day,
50 mg/kg,
i.p.
Male C57BL6 miceOBXSPTElevated sucrose preference[146]
Acute,
10, 30, 45, 60 nmol,
vmPFC microinjection
Male Wistar rats-FSTDecreased immobility[147]
rTMS: repetitive transcranial magnetic stimulation; CUMS: chronic unpredictable mild stress; CMS: chronic mild stress; ELS: early life stress; CHPG: (RS)-2-chloro-5-hydroxyphenylglycine; TST: Tail suspension test; i.c.v.: intracerebroventricular; CMS: chronic mild stress; HIPP: hippocampus; NMRI: naval medical research institute; CCI: chronic constriction injury; NP: neuropathic pain; SPT: sucrose preference test; SaPT: saccharine preference test; SD: Sprague–Dawley; ACEA: arachidonyl-2-chloroethylamide; WKY: Wistar Kyoto; FSL: Flinders Sensitive Line; i.v.: intravenous; OBX: olfactory bulbectomy.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bright, U.; Akirav, I. Modulation of Endocannabinoid System Components in Depression: Pre-Clinical and Clinical Evidence. Int. J. Mol. Sci. 2022, 23, 5526. https://doi.org/10.3390/ijms23105526

AMA Style

Bright U, Akirav I. Modulation of Endocannabinoid System Components in Depression: Pre-Clinical and Clinical Evidence. International Journal of Molecular Sciences. 2022; 23(10):5526. https://doi.org/10.3390/ijms23105526

Chicago/Turabian Style

Bright, Uri, and Irit Akirav. 2022. "Modulation of Endocannabinoid System Components in Depression: Pre-Clinical and Clinical Evidence" International Journal of Molecular Sciences 23, no. 10: 5526. https://doi.org/10.3390/ijms23105526

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