Therapeutic Effects of Cannabidiol on Methamphetamine Abuse: A Review of Preclinical Study

authors:

avatar Yasaman Razavi a , avatar Fariborz Keyhanfar b , avatar Ronak Shabani a , avatar Abbas Haghparast c , * , avatar Mehdi Mehdizadeh d , *

Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
Department of Pharmacology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
Reproductive Sciences and Technology Research Center, Department of Anatomy, Iran University of Medical Sciences, Tehran, Iran.
Corresponding Authors:

how to cite: Razavi Y, Keyhanfar F, Shabani R, Haghparast A, Mehdizadeh M. Therapeutic Effects of Cannabidiol on Methamphetamine Abuse: A Review of Preclinical Study. Iran J Pharm Res. 2021;20(4):e124546. https://doi.org/10.22037/ijpr.2021.114918.15106.

Abstract

As a strong and addictive psychostimulant, methamphetamine (METH) is often misused worldwide. Although relapse is the greatest challenge to the effective treatment of drug dependency, now, for METH addiction, there is not available accepted pharmacotherapy. To characterize a probable new target in this indication, a biological system comprised of endocannabinoids, known as the endocannabinoid system (ECS), has been advised. As a non-psychotomimetic Phytocannabinoid in Cannabis sativa, cannabidiol (CBD) has been used in preclinical and clinical studies for treating neuropsychiatric disorders. In this review article, we focus on the effects of CBD in the treatment of addiction in a preclinical investigation concerning the pharmaceutic effectiveness and the underlying mechanisms of action on drug abuse specially METH. Growing evidence shows that CBD is a potential therapeutic agent in reducing drug reward, as evaluated in conditioned place preference (CPP), brain-stimulation reward paradigms, and self- administration. Furthermore, CBD plays an effective role in decreasing relapse in animal research. Through multiple-mechanisms, there is a belief that CBD modulates brain dopamine responding to METH, resulting in a reduction of METH-seeking behaviors. As our studies indicate, CBD can decrease METH addiction-associated problems, for example, symptoms of withdrawal and craving. It is needed for conducting more preclinical investigations and upcoming clinical trials to entirely assess the CBD capability as interference for METH addiction.

Introduction

While stimulants such as cocaine, amphetamine, and methamphetamine (METH) are some of the most used forbidden recreational drugs worldwide (1, 2), no established medication was found for using in treating stimulant use disorders as yet (3). Long-lasting METH abuse, even following the abstinence period, may bring about cognitive deficits (4, 5). Imaging studies of long-lasting abusers show decreases in the density of brain dopamine and dopamine transporter (4-6) and the density of serotonin transporter and decreases in brain serotonin (7). If there are opiates, although these treatment approaches, together with methadone, have bettered the outcome of drug dependence in opiate-dependent people, opiate craving in all patients cannot be successfully stopped through these treatment approaches. Considering this situation, new molecules need to be discovered for treating disorders of drug abuse without any successful treatment approach.

During the past years, the enhanced identification of neural mechanisms involved in addictive disorders has been made available by the introduced modern knowledge and tools. The glutamatergic and dopaminergic systems are highly involved in the reinforcing outcomes of drugs and lengthy risk of relapse. Furthermore, the endocannabinoid system (ECBS) has an effect on the attainment and preservation of drug-seeking behaviors due to its function in reward and brain plasticity. According to the preliminary evidence, cannabidiol (CBD) can have healing outcomes for treating disorders of drug abuse (7-9). This review is intended to refine our present understanding of how CBD affects drug abuse such as METH and to discuss the existing data pointing to the possible effectiveness of CBD-based treatments for addiction treatment.

Cannabidiol pharmacology

CBD is considered a non-intoxicating constituent in the company of more than 80 different cannabinoids that are present only in the cannabis plant (8, 9). THC is the cannabinoid most closely associated with euphoria, dependence, and mental health side effects associated with cannabis consumption (10-12). CBD, which is another main cannabinoid in the Cannabis plant, did not have THC psychoactivity (10, 13). CBD holds a complicated pharmacodynamic profile, as a whole, as we know to interact with an extensive type of molecular targets. Although no full investigation has been done on all these targets, it is widely believed that almost all of them (49%) are enzymatic, 20% are the membrane and cellular transporters, 15% are receptors, and 15% are ion channels (15).

According to the reports of human studies, CBD has sedative, anxiolytic, antidepressant, mood stabilizer, and anti-craving properties (16-19). Lately, due to the anti-inflammatory and antioxidative (20) neuroprotective (21) properties of CBD and its hindrance to the rewarding effects of morphine (8), researchers have paid attention to it. The CBD effect of drug dependency has been explored by some studies. For example, the attenuated motivation of CBD to self-administration and relapse to METH (14). According to Trigo et al. ’s study, CBD prevents the potential relapse in cannabis dependency (15). Furthermore, the potential of CBD in decreasing susceptibility to drug addiction and relapse has been shown (16). In Razavi et al. ’s study, chronic ICV administration of CBD on impairments generated by METH in cognitive functions and recognition memory in mice chronically exposed to METH for the period of the abstinence (17) discovered the ability of CBD treatment for restoring spatial memory deficits. As these data denote, the rewarding effect and memory impairment of drug abuse is lessened by CBD treatment, suggesting a probable potential for combating relapse to drug-seeking.

Effects of Cannabidiol on reward circuitry

According to some data, numerous neuronal circuits concerned with drug addiction are modulated by CBD. CBD blocks the brain’s reward system. Recent evidence demonstrated that high doses of CBD (10 and 20 mg/kg) significantly increased Intracranial Self-Stimulation (ICSS) threshold frequency in the medial forebrain bundle (9). This possibly will denote the anti-reward effect of CBD; nevertheless, in the mentioned study, a 5 mg/kg dosage CBD, although efficient to reduce the morphine effects, did not adjust the acute strengthening properties of cocaine (8); it is also consistent with a previous report suggesting that CBD does not induce conditioned place preference (CPP) and therefore lacks hedonic properties (18). Several animal works have studied that single or repeated CBD administration can decrease the rewarding effects of cocaine and METH. CBD can decrease drug intake and weaken relapse to drug-seeking behavior (14, 19-21).

Furthermore, as a result of a rise in the expression level of hippocampal CB1Rs and BDNF, the repeated CBD administration not only decreases cocaine intake but also results in lasting neuroplasticity of the mesolimbic system (27). Unlike THC, CBD does not produce psychotomimetic properties and abuse potential (29), making it a promising candidate for future clinical use. Altogether, the CBD capability for reversing the raised movement of the mesolimbic DA reward system caused by the susceptibility to drug abuse can be a very important mechanism underlying its utility versus the addiction to psychostimulants and other drugs.

Effect of Cannabidiol in addictions

Drug addiction, known as mandatory drug-seeking, is a chronic condition that contains interchange drug withdrawal and relapse periods (30). Relatively few studies have examined the CBD effects on addiction and substance abuse in animals. CBD was examined for its anti-addictive properties in several animal models of cannabis, psychostimulants, opioid, alcohol, and nicotine addictions. Several studies have evaluated the CBD effect on drug dependency. For instance, previous studies have found the potentially relapse‐preventing effects of Sativex (THC/CBD) in cannabis dependency (23). In addition, CBD inhibits the reinstatement of cocaine and prevents the reinstatement of METH-induced CPP in rats (31).A recent study demonstrated that CBD treatment could prevent the reinstatement of Methylphenidate-induced CPP and produced shorter extinction latencies (22) . In an experiment with cocaine, the effects of a 10 mg/kg dose of CBD were evaluated on acquisition, consolidation, reconsolidation, extinction, and drug-primed reinstatement of cocaine using the drug CPP model, and the impact of CBD was examined in adult male mice. The results showed that CBD decreased preference of cocaine 20 days next treatment interruption, although there was no CBD effect on extinction, reconsolidation, or reinstatement of cocaine memory. These findings indicate that an acute 10 mg/kg dose of CBD has specific effects on cocaine memory processes.

Moreover, it has been suggested that CBD can be utilized as an efficient and innovative therapy for destabilizing the memories connected with drugs triggering abuse, thus lessening the drug relapse risk (32). Some studies were indicated evaluating the CBD effects on alcohol drinking associated with relapse and addiction (28, 33), for example, the CBD effects on motivation for drinking alcohol (28) using the alcohol self-administration paradigm (34). These findings indicate that the strengthening properties, motivation, and relapse for the consumption of ethanol were diminished by the CBD administration, suggesting the ability of CBD for treating disorders of alcohol use. While significant preclinical data on opioid drugs and CBD in the animal are accruing, the consistent findings of opioid abuse show that that CBD diminishes symptoms of morphine withdrawal (35, 36), and even in combination with THC, CBD can reduce the abstinence scores even higher than THC alone (35, 36). In addition, acute CBD lessened cue-caused reinstatement of heroin seeking examined one day following injection (26). These studies prove that CPP is not promoted by CBD (37, 38), or the reinforcing efficacy of brain stimulation is not increased (8), which are both definitive characteristics of addictive substances. CBD interrelates with neurotransmitter systems, which are essential for the effects of opioids and psychostimulants. For instance, CBD can allosterically regulate δ and μ opioid receptors (23) and cannot inhibit uptake in striatal dopamine synapses (24). Previous research revealed a reversed decline in expressing intra-accumbal AMPA glutamate receptor GluA1 subunits in heroin-trained mice after treating with CBD (19). According to the extraordinary speculation, the augmented cocaine-seeking for the abstinence period is partly reliant on the moderately augmented expression of GluA1 subunits in the nucleus accumbens (NAc) (19, 5). Thus, CBD may have high potential as an adjunct to cue exposure therapies for disorders, such as addiction.

Effect of Cannabidiol in METH abuse

For the implication of CBD on each phase of psychostimulant addiction, there are a few animal studies. The expansion of pharmacotherapies for treating stimulant use disorder has been in precedence in the studies conducted on addiction for more than 20 years, but the Food and Drug Administration (FDA) in the United States of America, or similar organizations in other countries did not have approved medication for this disease yet (42). The increased dopamine levels of the brain are the main mechanism of action related to the euphoria and abuse potential of METH; besides, as it can be suggested by a rising preclinical and clinical literature, blockers of dopamine uptake and releasers (methylphenidate, d-amphetamine, bupropion, modafinil, and methamphetamine) can be successful in the treatment of stimulant abuse and dependence (3). On the other hand, there is significant abuse and diversion potential for methylphenidate and d-amphetamine, and it seems that bupropion and modafinil have restricted clinical efficacy (3, 26). Consequently, to assist people with challenging stimulant use who are seeking treatment, we need new approaches. CBD inhibits the dopamine uptake, but this effect’s pharmacological relevance in a human being is unidentified (27, 28). CBD was not inherently hedonistic (18). Recently the 80 mg/kg CBD effects on reducing the motivation to self-administer METH and diminished methamphetamine-primed relapse to methamphetamine-seeking behavior following extinction were investigated in an animal study (14). Alternatively, according to Parker et al. ’s study, THC and CBD potentiate the extinction of CPP learning provoked by amphetamine in the rats (18), and a CB1 receptor antagonist did not reverse this effect, which implies that additional neurochemical mechanisms possibly will be involved. Furthermore, as Karimiet al. ’s study showed, the METH-induced reinstatement in extinguished rats could be suppressed by the ICV administration of the 10 μg/5 μL CBD through alteration of gene expression of cytokines, including interleukin-1β, -6, -10 and TNF-α (29, 30). These cytokines are recognized to regulate the neuronal activities of monoamine neurons which include DA neurons in a straight line by using activating cytokine receptors placed on DA and other monoamine neurons and not directly through the release of neuroactive molecules from glia cells (31, 32). Therefore, it was pointed out that METH re-exposure increases the expression of proinflammatory cytokines such as IL-1β and TNF-α, which leads to the release of neurotransmitters concerned with the METH reinstatement. It turns out that CBD worsens this type of METH-provoked neuroplasticity in the mesocorticolimbic system of DA.

Mechanisms of specific receptors underlying CBD’s activity versus METH addiction-associated behaviors remain unreported yet. Data have exposed that the CBD administration into NAc can prevent METH-stimulated behavioral sensitization and hyperlocomotion and by regulation of downstream phosphorylation of the mTOR/p70S6 kinase signaling pathway inside the NAc shell (33). Moreover, animals conditioned with METH demonstrate CPP related to upregulation of the Sigma1 receptor and several intracellular molecules, e.g., CREB and p-CREB, p-GSK-3β AKT, p-AKT, and GSK-3β in the hippocampus, ventral tegmental area (VTA), prefrontal cortex (PFC), and the NAc. CBD inhibits CPP induced by METH in a fashion dependent on the dose. The expression levels of Sigma1R, p-AKT, p-GSK3β, and p-CREB were enhanced significantly in the CPP stimulated by METH (34). These results propose that CBD can opposite some of the METH-brought neuroplastic changes and may have therapeutic potential on METH-driven behaviors. In general, CBD studies possibly will suggest too much effect in addictive behaviors of psychostimulants in the relapse phase and seems not to be found on rewarding effects. According to a study recently conducted, the hyperactivity and behavioral sensitization induced by amphetamine were reversed by administering a dosage of 100 ng/0.5 μL CBD into the NAc (33). Undeniably, for opposing METH-seeking and craving, CBD not only can be a valuable agent but its antipsychotic efficacy (35), taking into consideration that psychosis is a pervasive challenge in heavy METH addicts, make an additional valuable feature available. Table 1 shows a summary of preclinical researches on the CBD effects in animals that were exposed to METH (36-38).

Cannabidiol potential mechanisms of action

While many possible mechanisms have been proposed, the CBD action mode is not completely comprehended yet. Similar to other cannabinoids, bell-shaped dose-response curves are produced by CBD, and diverse mechanisms can act as a result of its concentration or the concurrent existence of other cannabinoid ligands. The action of serotonin 5HT1A receptor, peroxisome proliferator-activated receptor-gamma (PPARγ), the metabotropic CB1 and CB2 receptors, and members of the TRPV family can be directly or indirectly regulated by CBD (39, 40). As indicated by recent evidence, withdrawal signs of METH-dependence may be reduced by acute administration of CBD, but the treatment must be elongated over time to facilitate METH quitting. Mostly hypothetically, the CBD ability in treating addictive disorders has been associated with modulating endocannabinoid, serotoninergic and glutamatergic systems (Figure 1).

Cannabinoid receptors

CBD includes a minimal affinity for CB1 and CB2, which are two recognized cannabinoid receptors. The ability of the CB1 agonists (WIN55212 and CP55940) to affect contractions at doses that were noticeably less than those of CBD required for activating cannabinoid receptors was attenuated by CBD (41, 42). As reported, CBD plays a role as an antagonist of CB1R agonists, for example, WIN-55212 and CP-55940 (43). Also, as CB1R internalization was inhibited by CBD (44), there is a hypothesis that the seen antagonistic activity may perhaps be based on negative allosteric modulation of CB1R instead of on orthosteric binding. According to the evidence obtained by these findings, in-vitro CBD acts as a non-competitive negative allosteric modulator of CB1R (41). More recently, a study reported that these actions are cannabinoid-receptor-mediated. Probably, CBD inhibiting fatty acid amide hydrolase (FAAH) activity increases the level of arachidonoylethanolamide (AEA), which actives CB1R (45). Moreover, CBD was determined to operate as a CB2R antagonist or inverse agonist (43). CBD includes a high-potency antagonist of cannabinoid-receptor agonists in the brain of mice and also in membranes from cells transfected with human CB2. Also, an inverse agonism is exhibited by CBD at the human CB2 receptor. Lots of the effects recorded with CBD, such as its anti-inflammatory properties, may be rationalized by these unexpected observations. Also, these findings report that CB2R antagonism attenuates CBD-produced neuroprotection (46).

The 5-HT1a Receptor

CBD has been recently found that activating post-synaptic 5-HT1A receptors will possibly apply anxiolytic effects in the crosstalk between cannabinoids and serotoninergic signaling (34). The CBD’s anxiolytic properties have been proven in different animal models, such as the elevated plus-maze and conditioned emotional response (32, 33).

The 5-HT1A receptors play essential parts in the pathophysiology of depression, anxiety, and aggression. The agonist [3H] 8-OHDPAT from the cloned human 5-HT1a receptor is dislocated by CBD in a concentration-dependent manner. Contrastingly, agonist from the receptor in the same micromolar concentration range is not displaced by the major psychoactive component of cannabis, THC. CBD is considered as a modest-affinity agonist at the human 5-HT1a receptor; on the other hand, CBD increases the agonist serotonin, as well as GTPgS binding in this G-protein-coupled receptor (GPCR) system. Furthermore, the cAMP concentration at similar apparent levels of receptor occupancy is decreased by both CBD and 5-HT in this GPCR system negatively coupled to cAMP production (47, 48). Besides, the anti-craving effect of CBD may be also contributed by the agonist activity of CBD in the direction of 5HT1A receptors; similarly, the substance abuse relapses by regulation of the stress management, anxiety symptoms, and drug reward system are reduced by the agonist action of CBD in the direction of 5HT1A receptors (16). Eventually, the glutamatergic signaling by modulating endocannabinoid and serotoninergic systems might be regulated by CBD, and since dysregulating the glutamatergic transmission has been broadly associated with both abuse relapses and drug-seeking behaviors (16), this mechanism may be also involved in treating addictive behaviors (49). In rats, anti- aversive outcomes in the raised plus-maze and flight-induced by local electric stimulation are created by CBD administration into the dorsal parts of periaqueductal gray matter (dPAG). WAY-100635, a 5HT1A antagonist prevented these effects (50){Campos, 2008, Involvement of 5HT1A receptors in the anxiolytic-like effects of cannabidiol injected into the dorsolateral periaqueductal gray of rats}. Also, it seems that the basal ganglia (51), the bed nucleus of stria terminallis (52), the prelimbic PFC (53), and the dorsal raphe nucleus (9, 47), which are other brain regions, mediate the effects of CBD through 5HT1A receptors. Not long ago, Magenet al. verified that activating 5-HT1A receptors positioned in forebrain regions that include the hippocampus, and also, recovered cognitive and locomotor function weakened by bile-duct ligation was induced by CBD (5 mg/kg, i.p) (54). As a whole, these data denote that CBD possibly activates the 5-HT1A receptor, resulting in improving cognitive and functional impairment.

A Potent Antioxidant

The well-known antioxidants are phenols, including resorcinols. Similarly, monophenols, plant cannabinoids, monophenolic ethers (like THC), or resorcinols (as CBD) are strong antioxidants. In a study, Hampsonet al. (64) observed that CBD is a non-psychoactive ingredient of marijuana; also, it has a stronger effect than either α-tocopherol, which contains vitamin E, and is a dietary antioxidant, or ascorbate, which contains vitamin C, could prevent ROS-caused cell death and glutamate neurotoxicity. In a newer study (15), Hamelinket al. discovered that CBD safeguarded rats as opposed to hippocampal- entorhinal-cortical neurodegeneration while they were administered simultaneously with ethanol exposure. As they also have shown, this safeguard was not a result of NMDA-receptor antagonism, in the same way as other antioxidant NMDA antagonists, did not stop cell death and attached the CBD activity to its antioxidative impacts. By the original 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical method explained by Brand-Williamset al., CBD and THC were assessed in another study for antioxidant activity (57). Also, according to another study, CBD revealed stronger antioxidative ability than THC (55). These data indicate that CBD can be a possibly useful therapeutic agent for treating oxidative neurological conditions.

PPARγ

It was pointed out that CBD binds to the peroxisome proliferator-activated receptor-gamma (PPARγ) as it is supposed that the glitazone receptor is responsible for lipid storage and glucose metabolism (56, 57). PPARγ not only regulates inflammatory responses but also regulates the expression of genes associated with lipid and glucose homeostasis. Consequently, detected disorders of glucose metabolism and immune/inflammatory processes by PPARγ activation may be ameliorated by CBD (58), and it is supposed that some anticancer effects of CBD are mediated through interaction with PPARγ. CBD has also inhibited tumor cell viability.

TRPV1 and the Effects of Cannabidiol

Also, at least in some cases, CBD and other non-psychotomimetic phytocannabinoids are able to act through the transient receptor potential vanilloid (TRPV), which is a member of the ion channel receptor family. TRPV1 in vitro is activated and desensitized by CBD and cannabidivarin (CBDV) (59). Activation of TRPV1 receptors plays a role in the bell-formed dose-response curve of the anxiolytic activity of CBD. Treatment of animals with a TRPV1 antagonist prevented the absence of effects seen at elevated dosages of CBD (60). Also, it seems that TRPV1 plays a part in the anti hyperalgesic effects of CBD (61) besides in CBD effects on the disruption of sensorimotor gating aroused from NMDA antagonists (62). Evidence indicated that TRPV1 channels are on striatal GABAergic neurons and neurons of glutamatergic in the frontal cortex(63). It is suggested that CBD via TRPV1 may modify GABA release and brain glutamate, which leads to decreased cocaine reward. (64). Previous research showed that pretreatment with an antagonist of TRPV1(capsazepine) was in a position to stop CBD- induced reduction in the cocaine self-administration (65), proposing that activation of TRPV1 might as well come to the aid of the therapeutic effects from CBD.

Neuroprotective effects as a selective therapeutic by Cannabidiol

An essential system of action of neuropsychiatric medications to maintain the functions and structure of neural cells is constituted by neuroprotection, which promotes protection against protein aggregation, excitotoxicity, organelles damage, inflammation, and oxidative stress (66). It was proposed that by some intrinsic pharmacological properties, CBD impedes the THC effects. When CBD is administered alone, its hypnotic, anticonvulsive, neuroprotective, and hormonal effects, i.e., the increased corticosterone and cortisol levels, are produced spontaneously and support the hypothesis that CBD might have anxiolytic and/or antipsychotic effects (77). However, as Niesink and van Laar (10) verified, CBD did not have more or less effective in normal physiological processes. In order to be able to express the effect of CBD, the natural “tone” of the endocannabinoid system should be disrupted by a stimulus such as pain or a shock reaction, or another cannabinoid such as THC (67).

An early study linking CBD to neuroprotection shows that CBD plays an antioxidant role against toxicity and/or oxidative stress generated by amphetamine (68, 69). The neuroprotective effects of CBD may perhaps also consist of neuroinflammatory mechanisms. Ab-induced neuroinflammation is diminished by CBD; also, hippocampal neurogenesis is promoted by CBD. It seems that these effects occur partially by activating receptors of PPARγ (70). Furthermore, neuroprotective effects in the cerebral malaria model are exhibited by CBD. Cognitive function rescue is promoted by CBD treatment with an increase in the expression of BDNF and decreased proinflammatory cytokines (TNF-α) and (IL-6) levels (71). In animal encephalopathy models, motor activity restored levels of 5-HT and BDNF, and cognition were improved by CBD by activating the 5HT1A receptor (72). While studies linking CBD to autophagy in neuropsychiatric conditions in quantity are insufficient, this process can be modulated by CBD (73, 74). Anticonvulsant effects associated with the activation of the hippocampal autophagy path in the chronic stage of pilocarpine-stimulated seizure were produced by CBD explicitly in the brain (75).

Potential mechanisms underlying CBD’s action and the main molecular targets. CBD inhibit, the enzyme which metabolizes anandamide i.e. FAAH. and activate CB1 and/or CB2 receptors indirectly. Also CBD may act as a CB1R negative allosteric modulator, a CB2R partial agonist or antagonist/inverse agonist. CBD activates the transient receptor potential channels (TRPV1), 5‐HT1A receptor and as antagonist of the receptor GPR55.Also promote PPARγ receptors, increased anti-inflammatory cytokines responses resulting in neuroprotection
Table 1

Effects of CBD in animals exposed to METH

Evaluation methoddose of CBDMain resultsReferences
CPP5 mg/kg. i.p.CBD potentiates the extinction of Amphetamine-induced CPP and this effect is not reversed by CB1 receptor antagonistParker et al. 2004
Psychomotor sensitization100 ng/0.50 μLCBD attenuates Amphetamine-induced sensitization in nucleus accumbens shell. CBD controls downstream phosphorylation of the mTOR/p70S6 kinase signaling pathways directly within the shell of NAc.Renard et al. 2016
Self-administration20, 40, and 80; i.p. mg/kgCBD decreases the motivation to self-administer METH and reduces METH-primed relapse after extinction.Hay et al. 2018
CPP10 μg/5 μLICV microinjection of CBD supress the METH-induced reinstatement even in REM sleep deprived rats.Karimi et al. 2018
CPP10 μg/5 μLCBD treatment reduced the mRNA expression of cytokines in the PFC and HIP. Also, CBD treatment before REM sleep deprivation augments the TNF‐α, IL‐1β, IL‐6, and IL‐10 levels in the HIP but diminishes IL‐10 in the PFC.Karimi et al. 2020
CPP10, 20, 40, and 80; mg/kg i.p.CBD prevent METH-induced CPP and causes differential inhibitory responses in the cellular protein abundance of, p-AKT, Sigma1R, p-GSK3β, and p-CREB across various brain regions.Yang et al. 2020
Chronic exposure10 and 50 μg/5 μLICV microinjection of CBD improves spatial memory and reverses short- and long-term memory that are impaired by chronic exposure of METH during abstinenceRazavi et al. 2020
Chronic exposure10 and 50 μg/5 μLICV administration of CBD enhance the mRNA expression levels of BDNF/TrkB; RAF1, and NGF/TrkA in the HIP during abstinence.Razavi et al. 2021
CPP10, 50, 100, and 200 μgICV administration of CBD shifted the establishment of METH-induced CPP toward a lower dose. Concurrent CBD and METH treatments during sensitization phase established METH-induced CPP with sub-threshold doses of METH.Khaneghini et al. 2021
CPP10 and 50 µgIntra-CA1 microinjection of SCH23390 impairs CBD’s suppressive impact on both acquisition and expression phases of METH-induced CPPAnooshe et al. 2021
CPP10 and 50 µgCBD reduce METH-induced CPP. Intra-CA1 microinjection of sulpiride reversed the decreasing effects of CBD on METH-induced CPP in both acquisition and expression phases but more prominent in the expression phaseHassanlou et al. 2021

Conclusion

To sum up, currently, we have inadequate evidence of the CBD’s possible therapeutic advantages of METH abuse and its frequently associated adverse symptoms. The quantity smallness of human research and the absence of clinical trials caused an apparent literature inadequacy. In preclinical research, CBD indicates pharmacological effectiveness in lessening propensity to relapse and drug reward. As reviewed, the action mechanisms of CBD are highly intricate and concerned with multiple receptors. Pharmacological (in-vivo) studies display that TRPV1, 5-HT1A, CB1, and CB2 receptors are essentially concerned with the mechanism of actions of the CBD. The mentioned receptors may modulate the activity of DA neurons in the DA and VTA release directly or in the NAc indirectly. Therefore, the mesolimbic DA system was able to operate as an ultimate target of anti-addiction effects of underlying CBD. There is an obvious necessity for other preclinical studies and forthcoming clinical trials to completely assess the CBD ability as an intervention therapy for METH addictive conditions. As a potential agent for the treatment of human addictive behaviors, evaluation of CBD should be more carefully carried out. They also confirm its low addictive risk as a new intervention for addiction as well as data that indicate CBD is not supporting on its own. For translating research findings into clinical settings, validating the CBD efficacy and safety in lessening the craving and relapse will be necessary for clinical and preclinical trials.

Conflict of interest

The authors declare that there is no conflict of interest.

Funding

We declare that there was no source of funding.

References

  • 1.

    van Amsterdam J, Pennings E, Brunt T, van den Brink W. Physical harm due to chronic substance use. Regul. Toxicol. Pharmacol. 2013;66:83-7. [PubMed ID: 23542091].

  • 2.

    Hall W, Degenhardt L. The adverse health effects of chronic cannabis use. Drug Test. Anal. 2014;6:39-45. [PubMed ID: 23836598].

  • 3.

    Haile CN, Mahoney III JJ, Newton TF, De La Garza II R. Pharmacotherapeutics directed at deficiencies associated with cocaine dependence: focus on dopamine, norepinephrine and glutamate. Pharmacol. Ther. 2012;134:260-77. [PubMed ID: 22327234].

  • 4.

    Baicy K, London ED. Corticolimbic dysregulation and chronic methamphetamine abuse. Addiction. 2007;102:5-15.

  • 5.

    Meredith CW, Jaffe C, Ang-Lee K, Saxon AJ. Implications of chronic methamphetamine use: a literature review. Harv. Rev. Psychiatry. 2005;13:141-54. [PubMed ID: 16020027].

  • 6.

    Barr AM, Panenka WJ, MacEwan GW, Thornton AE, Lang DJ, Honer WG, Lecomte T. The need for speed: an update on methamphetamine addiction. J. Psychiatry Neurosci. 2006.

  • 7.

    Zuardi AW. Cannabidiol: from an inactive cannabinoid to a drug with wide spectrum of action. Braz. J. Psychiatry. 2008;30:271-80. [PubMed ID: 18833429].

  • 8.

    Mechoulam R, Carlini EA. Toward drugs derived from cannabis. Naturwissenschaften. 1978;65:174-9. [PubMed ID: 351429].

  • 9.

    Katsidoni V, Anagnostou I, Panagis G. Cannabidiol inhibits the reward‐facilitating effect of morphine: involvement of 5‐HT1A receptors in the dorsal raphe nucleus. Addict. Biol. 2013;18:286-96. [PubMed ID: 22862835].

  • 10.

    Niesink RJ, van Laar MW. Does Cannabidiol Protect Against Adverse Psychological Effects of THC? Front. Psychiatry. 2013;4:130.

  • 11.

    Carbuto M, Sewell RA, Williams A, Forselius-Bielen K, Braley G, Elander J, Pittman B, Schnakenberg A, Bhakta S, Perry E, Ranganathan M, D’Souza DC. The safety of studies with intravenous Δ 9-tetrahydrocannabinol in humans, with case histories. Psychopharmacology. 2012;219:885-96. [PubMed ID: 21845389].

  • 12.

    Maldonado R, Berrendero F, Ozaita A, Robledo P. Neurochemical basis of cannabis addiction. Neuroscience. 2011;181:1-17. [PubMed ID: 21334423].

  • 13.

    Morgan CJ, Freeman TP, Schafer GL, Curran HV. Cannabidiol attenuates the appetitive effects of Δ 9-tetrahydrocannabinol in humans smoking their chosen cannabis. Neuropsychopharmacology. 2010;35:1879-85. [PubMed ID: 20428110].

  • 14.

    Hay GL, Baracz SJ, Everett NA, Roberts J, Costa PA, Arnold JC, McGregor IS, Cornish JL. Cannabidiol treatment reduces the motivation to self-administer methamphetamine and methamphetamine-primed relapse in rats. J. Psychopharmacol. 2018;32:1369-78. [PubMed ID: 30260267].

  • 15.

    Trigo JM, Soliman A, Staios G, Quilty L, Fischer B, George TP, Rehm J, Selby P, Barnes AJ, Huestis MA, Foll BL. Sativex associated with behavioral-relapse prevention strategy as treatment for cannabis dependence: a case series. J. Addict. Med. 2016;10:274.

  • 16.

    Prud’homme M, Cata R, Jutras-Aswad D. Cannabidiol as an intervention for addictive behaviors: a systematic review of the evidence. Subst. Abuse: Res. Treat. 2015;9:33-8.

  • 17.

    Razavi Y, Shabani R, Mehdizadeh M, Haghparast A. Neuroprotective effect of chronic administration of cannabidiol during the abstinence period on methamphetamine-induced impairment of recognition memory in the rats. Behav. Pharmacol. 2020;31:385-96. [PubMed ID: 32032100].

  • 18.

    Parker LA, Burton P, Sorge RE, Yakiwchuk C, Mechoulam R. Effect of low doses of delta9-tetrahydrocannabinol and cannabidiol on the extinction of cocaine-induced and amphetamine-induced conditioned place preference learning in rats. Psychopharmacology (Berl). 2004;175:360-6. [PubMed ID: 15138755].

  • 19.

    Ren Y, Whittard J, Higuera-Matas A, Morris CV, Hurd YL. Cannabidiol, a nonpsychotropic component of cannabis, inhibits cue-induced heroin seeking and normalizes discrete mesolimbic neuronal disturbances. J. Neurosci. 2009;29:14764-9. [PubMed ID: 19940171].

  • 20.

    Luján MÁ, Castro-Zavala A, Alegre-Zurano L, Valverde O. Repeated Cannabidiol treatment reduces cocaine intake and modulates neural proliferation and CB1R expression in the mouse hippocampus. Neuropharmacology. 2018;143:163-75. [PubMed ID: 30273593].

  • 21.

    Viudez‐Martínez A, García‐Gutiérrez MS, Navarrón CM, Morales‐Calero MI, Navarrete F, Torres‐Suárez AI, Manzanares J. Cannabidiol reduces ethanol consumption, motivation and relapse in mice. Addict. Biol. 2018;23:154-64. [PubMed ID: 28194850].

  • 22.

    Kashefi A, Tomaz C, Jamali S, Rashidy-Pour A, Vafaei AA, Haghparast A. Cannabidiol attenuated the maintenance and reinstatement of extinguished methylphenidate-induced conditioned place preference in rats. Brain Res. Bull. 2021;166:118-27. [PubMed ID: 33264654].

  • 23.

    Kathmann M, Flau K, Redmer A, Tränkle C, Schlicker E. Cannabidiol is an allosteric modulator at mu-and delta-opioid receptors. Naunyn Schmiedebergs Arch. Pharmacol. 2006;372:354-61. [PubMed ID: 16489449].

  • 24.

    Pandolfo P, Silveirinha V, dos Santos-Rodrigues A, Venance L, Ledent C, Takahashi RN, Cunha RA, Köfalvi A. Cannabinoids inhibit the synaptic uptake of adenosine and dopamine in the rat and mouse striatum. Eur. J. Pharmacol. 2011;655:38-45. [PubMed ID: 21266173].

  • 25.

    Conrad KL, Ford K, Marinelli M, Wolf ME. Dopamine receptor expression and distribution dynamically change in the rat nucleus accumbens after withdrawal from cocaine self-administration. Neuroscience. 2010;169:182-94. [PubMed ID: 20435100].

  • 26.

    W Stoops W, R Rush C. Agonist replacement for stimulant dependence: a review of clinical research. Curr. Pharm. Des. 2013;19:7026-35. [PubMed ID: 23574440].

  • 27.

    Bergamaschi MM, Queiroz RH, Zuardi AW, Crippa JA. Safety and side effects of cannabidiol, a Cannabis sativa constituent. Curr. Drug Saf. 2011;6:237-49. [PubMed ID: 22129319].

  • 28.

    Zhornitsky S, Potvin S. Cannabidiol in humans-the quest for therapeutic targets. Pharm. (Basel). 2012;5:529-52.

  • 29.

    Karimi-Haghighi S, Haghparast A. Cannabidiol inhibits priming-induced reinstatement of methamphetamine in REM sleep deprived rats. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2018;82:307-13. [PubMed ID: 28870635].

  • 30.

    Karimi-Haghighi S, Dargahi L, Haghparast A. Cannabidiol modulates the expression of neuroinflammatory factors in stress- and drug-induced reinstatement of methamphetamine in extinguished rats. Addict. Biol. 2020;25:e12740. [PubMed ID: 30793820].

  • 31.

    Palazzolo DL, Quadri SK. Interleukin-1 stimulates catecholamine release from the hypothalamus. Life Sci. 1990;47:2105-9. [PubMed ID: 2266781].

  • 32.

    Shintani F, Kanba S, Nakaki T, Nibuya M, Kinoshita N, Suzuki E, Yagi G, Kato R, Asai M. Interleukin-1 beta augments release of norepinephrine, dopamine, and serotonin in the rat anterior hypothalamus. J. Neurosci. 1993;13:3574-81. [PubMed ID: 8393485].

  • 33.

    Renard J, Loureiro M, Rosen LG, Zunder J, de Oliveira C, Schmid S, Rushlow WJ, Laviolette SR. Cannabidiol Counteracts Amphetamine-Induced Neuronal and Behavioral Sensitization of the Mesolimbic Dopamine Pathway through a Novel mTOR/p70S6 Kinase Signaling Pathway. J. Neurosci. 2016;36:5160-9. [PubMed ID: 27147666].

  • 34.

    Yang G, Liu L, Zhang R, Li J, Leung CK, Huang J, Li Y, Shen B, Zeng X, Zhang D. Cannabidiol attenuates methamphetamine-induced conditioned place preference via the Sigma1R/AKT/GSK-3β/CREB signaling pathway in rats. Toxicol. Res. Camb;(2020) 9:202-11.

  • 35.

    Leweke FM, Piomelli D, Pahlisch F, Muhl D, Gerth CW, Hoyer C, Klosterkötter J, Hellmich M, Koethe D. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl. Psychiatry. 2012;2:e94. [PubMed ID: 22832859].

  • 36.

    Khanegheini A, Khani M, Zarrabian S, Yousefzadeh-Chabok S, Taleghani BK, Haghparast A. Cannabidiol enhanced the development of sensitization to the expression of methamphetamine-induced conditioned place preference in male rats. J. Psychiatr. Res. 2021;137:260-5. [PubMed ID: 33725638].

  • 37.

    Hassanlou AA, Jamali S, RayatSanati K, Mousavi Z, Haghparast A. Cannabidiol modulates the METH-induced conditioned place preference through D2-like dopamine receptors in the hippocampal CA1 region. Brain Res. Bull. 2021;172:43-51. [PubMed ID: 33862125].

  • 38.

    Anooshe M, Nouri K, Karimi-Haghighi S, Mousavi Z, Haghparast A. Cannabidiol efficiently suppressed the acquisition and expression of methamphetamine-induced conditioned place preference in the rat. Behav. Brain Res. 2021;404:113158. [PubMed ID: 33571569].

  • 39.

    Campos AC, Moreira FA, Gomes FV, Del Bel EA, Guimarães FS. Multiple mechanisms involved in the large-spectrum therapeutic potential of cannabidiol in psychiatric disorders. Philos. Trans. R. Soc. Lond. B: Biol. Sci. 2012;367:3364-78. [PubMed ID: 23108553].

  • 40.

    Ligresti A, De Petrocellis L, Di Marzo V. From phytocannabinoids to cannabinoid receptors and endocannabinoids: pleiotropic physiological and pathological roles through complex pharmacology. Physiol. Rev. 2016;96:1593-659. [PubMed ID: 27630175].

  • 41.

    Laprairie RB, Bagher AM, Kelly ME, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br. J. Pharmacol. 2015;172:4790-805. [PubMed ID: 26218440].

  • 42.

    McPartland JM, Duncan M, Di Marzo V, Pertwee RG. Are cannabidiol and Δ(9) -tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. Br. J. Pharmacol. 2015;172:737-53. [PubMed ID: 25257544].

  • 43.

    Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG. Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br. J. Pharmacol. 2007;150:613-23. [PubMed ID: 17245363].

  • 44.

    Laprairie RB, Bagher AM, Kelly ME, Dupré DJ, Denovan-Wright EM. Type 1 cannabinoid receptor ligands display functional selectivity in a cell culture model of striatal medium spiny projection neurons. J. Biol. Chem. 2014;289:24845-62. [PubMed ID: 25037227].

  • 45.

    Premoli M, Aria F, Bonini SA, Maccarinelli G, Gianoncelli A, Della Pina S, Tambaro S, Memo M, Mastinu A. Cannabidiol: Recent advances and new insights for neuropsychiatric disorders treatment. Life Sci. 2019;224:120-7. [PubMed ID: 30910646].

  • 46.

    Castillo A, Tolón MR, Fernández-Ruiz J, Romero J, Martinez-Orgado J. The neuroprotective effect of cannabidiol in an in-vitro model of newborn hypoxic-ischemic brain damage in mice is mediated by CB(2) and adenosine receptors. Neurobiol. Dis. 2010;37:434-40. [PubMed ID: 19900555].

  • 47.

    Rock EM, Bolognini D, Limebeer CL, Cascio MG, Anavi-Goffer S, Fletcher PJ, Mechoulam R, Pertwee RG, Parker LA. Cannabidiol, a non-psychotropic component of cannabis, attenuates vomiting and nausea-like behaviour via indirect agonism of 5-HT(1A) somatodendritic autoreceptors in the dorsal raphe nucleus. Br. J. Pharmacol. 2012;165:2620-34. [PubMed ID: 21827451].

  • 48.

    Russo EB, Burnett A, Hall B, Parker KK. Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem. Res. 2005;30:1037-43. [PubMed ID: 16258853].

  • 49.

    Rodríguez-Muñoz M, Sánchez-Blázquez P, Merlos M, Garzón-Niño J. Endocannabinoid control of glutamate NMDA receptors: the therapeutic potential and consequences of dysfunction. Oncotarget. 2016;7:55840-62. [PubMed ID: 27323834].

  • 50.

    Campos AC, Guimarães FS. Involvement of 5HT1A receptors in the anxiolytic-like effects of cannabidiol injected into the dorsolateral periaqueductal gray of rats. Psychopharmacology (Berl). 2008;199:223-30. [PubMed ID: 18446323].

  • 51.

    Espejo-Porras F, Fernández-Ruiz J, Pertwee RG, Mechoulam R, García C. Motor effects of the non-psychotropic phytocannabinoid cannabidiol that are mediated by 5-HT1A receptors. Neuropharmacology. 2013;75:155-63. [PubMed ID: 23924692].

  • 52.

    Gomes FV, Resstel LB, Guimarães FS. The anxiolytic-like effects of cannabidiol injected into the bed nucleus of the stria terminalis are mediated by 5-HT1A receptors. Psychopharmacology (Berl). 2011;213:465-73. [PubMed ID: 20945065].

  • 53.

    Fogaça MV, Reis FM, Campos AC, Guimarães FS. Effects of intra-prelimbic prefrontal cortex injection of cannabidiol on anxiety-like behavior: involvement of 5HT1A receptors and previous stressful experience. Eur. Neuropsychopharmacol. 2014;24:410-9. [PubMed ID: 24321837].

  • 54.

    Twardowschy A, Castiblanco-Urbina MA, Uribe-Mariño A, Biagioni AF, Salgado-Rohner CJ, Crippa JA, Coimbra NC. The role of 5-HT1A receptors in the anti-aversive effects of cannabidiol on panic attack-like behaviors evoked in the presence of the wild snake Epicrates cenchria crassus (Reptilia, Boidae). J. Psychopharmacol. 2013;27:1149-59. [PubMed ID: 23926240].

  • 55.

    Hayakawa K, Mishima K, Nozako M, Ogata A, Hazekawa M, Liu AX, Fujioka, Abe K, Hasebe N, Egashira N, Iwasaki K, Fujiwara M. Repeated treatment with cannabidiol but not Delta9-tetrahydrocannabinol has a neuroprotective effect without the development of tolerance. Neuropharmacology. 2007;52:1079-87. [PubMed ID: 17320118].

  • 56.

    O’Sullivan SE, Sun Y, Bennett AJ, Randall MD, Kendall DA. Time-dependent vascular actions of cannabidiol in the rat aorta. Eur. J. Pharmacol. 2009;612:61-8. [PubMed ID: 19285060].

  • 57.

    Granja AG, Carrillo-Salinas F, Pagani A, Gómez-Cañas M, Negri R, Navarrete C, Mecha M, Mestre L, Fiebich BL, Cantarero I, Calzado MA, Bellido ML, Fernandez-Ruiz J, Appendino G, Guaza C, Muñoz E. A cannabigerol quinone alleviates neuroinflammation in a chronic model of multiple sclerosis. J. Neuroimmune. Pharmacol. 2012;7:1002-16. [PubMed ID: 22971837].

  • 58.

    Rajasekaran A, Venkatasubramanian G, Berk M, Debnath M. Mitochondrial dysfunction in schizophrenia: pathways, mechanisms and implications. Neurosci. Biobehav. Rev. 2015;48:10-21. [PubMed ID: 25446950].

  • 59.

    Iannotti FA, Hill CL, Leo A, Alhusaini A, Soubrane C, Mazzarella E, Russo E, Whalley BJ, Marzo VD, Stephens GJ. Nonpsychotropic plant cannabinoids, cannabidivarin (CBDV) and cannabidiol (CBD), activate and desensitize transient receptor potential vanilloid 1 (TRPV1) channels in-vitro: potential for the treatment of neuronal hyperexcitability. ACS Chem. Neurosci. 2014;5:1131-41. [PubMed ID: 25029033].

  • 60.

    Campos AC, Guimarães FS. Evidence for a potential role for TRPV1 receptors in the dorsolateral periaqueductal gray in the attenuation of the anxiolytic effects of cannabinoids. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2009;33:1517-21. [PubMed ID: 19735690].

  • 61.

    Costa B, Giagnoni G, Franke C, Trovato AE, Colleoni M. Vanilloid TRPV1 receptor mediates the antihyperalgesic effect of the nonpsychoactive cannabinoid, cannabidiol, in a rat model of acute inflammation. Br. J. Pharmacol. 2004;143:247-50. [PubMed ID: 15313881].

  • 62.

    Long LE, Malone DT, Taylor DA. Cannabidiol reverses MK-801-induced disruption of prepulse inhibition in mice. Neuropsychopharmacology. 2006;31:795-803. [PubMed ID: 16052245].

  • 63.

    Edwards JG. TRPV1 in the central nervous system: synaptic plasticity, function, and pharmacological implications. Prog Drug Res. 2014:77-104. [PubMed ID: 24941665].

  • 64.

    Bisogno T, Hanuš L, De Petrocellis L, Tchilibon S, Ponde DE, Brandi I, Moriello AS, Davis JB, Mechoulam R, Marzo VD. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br. J. Pharmacol. 2001;134:845-52. [PubMed ID: 11606325].

  • 65.

    Galaj E, Bi GH, Yang HJ, Xi ZX. Cannabidiol attenuates the rewarding effects of cocaine in rats by CB2, 5-HT1A and TRPV1 receptor mechanisms. Neuropharmacology. 2020;167:107740. [PubMed ID: 31437433].

  • 66.

    Filipović D, Todorović N, Bernardi RE, Gass P. Oxidative and nitrosative stress pathways in the brain of socially isolated adult male rats demonstrating depressive- and anxiety-like symptoms. Brain Struct. Funct. 2017;222:1-20. [PubMed ID: 27033097].

  • 67.

    Alger BE, Kim J. Supply and demand for endocannabinoids. Trends Neurosci. 2011;34:304-15. [PubMed ID: 21507493].

  • 68.

    Mecha M, Torrao AS, Mestre L, Carrillo-Salinas FJ, Mechoulam R, Guaza C. Cannabidiol protects oligodendrocyte progenitor cells from inflammation-induced apoptosis by attenuating endoplasmic reticulum stress. Cell Death Dis. 2012;3:e331. [PubMed ID: 22739983].

  • 69.

    Harvey BS, Ohlsson KS, Mååg JL, Musgrave IF, Smid SD. Contrasting protective effects of cannabinoids against oxidative stress and amyloid-β evoked neurotoxicity in-vitro. Neurotoxicology. 2012;33:138-46. [PubMed ID: 22233683].

  • 70.

    Scuderi C, Steardo L, Esposito G. Cannabidiol promotes amyloid precursor protein ubiquitination and reduction of beta amyloid expression in SHSY5YAPP+ cells through PPARγ involvement. Phytother. Res. 2014;28:1007-13. [PubMed ID: 24288245].

  • 71.

    Campos AC, Brant F, Miranda AS, Machado FS, Teixeira AL. Cannabidiol increases survival and promotes rescue of cognitive function in a murine model of cerebral malaria. Neuroscience. 2015;289:166-80. [PubMed ID: 25595981].

  • 72.

    Avraham Y, Grigoriadis N, Poutahidis T, Vorobiev L, Magen I, Ilan Y, Mechoulam R, Berry EM. Cannabidiol improves brain and liver function in a fulminant hepatic failure-induced model of hepatic encephalopathy in mice. Br. J. Pharmacol. 2011;162:1650-8. [PubMed ID: 21182490].

  • 73.

    Yang L, Rozenfeld R, Wu D, Devi LA, Zhang Z, Cederbaum A. Cannabidiol protects liver from binge alcohol-induced steatosis by mechanisms including inhibition of oxidative stress and increase in autophagy. Free Radic. Biol. Med. 2014;68:260-7. [PubMed ID: 24398069].

  • 74.

    Koay LC, Rigby RJ, Wright KL. Cannabinoid-induced autophagy regulates suppressor of cytokine signaling-3 in intestinal epithelium. Am. J. Physiol. Gastrointest. Liver Physiol. 2014;307:G140-8. [PubMed ID: 24833710].

  • 75.

    Hosseinzadeh M, Nikseresht S, Khodagholi F, Naderi N, Maghsoudi N. Cannabidiol post-treatment alleviates rat epileptic-related behaviors and activates hippocampal cell autophagy pathway along with antioxidant defense in chronic phase of pilocarpine-induced seizure. J. Mol. Neurosci. 2016;58:432-40. [PubMed ID: 26738731].

Sierra
2023-04-17 04:22:08
I have been addicted to meth for around 10 years about I know you say the research need more trials and studying and I pray it is done soon because there is a lot of meth addicts who need help with the withdrawals and cravings and feeling you get during use and after stopping use. I am one of those people who need help.