ijms-logo

Journal Browser

Journal Browser

Endocannabinoids, Cannabinoids and Psychiatry: Biological Mechanisms 2.0

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Pathology, Diagnostics, and Therapeutics".

Deadline for manuscript submissions: closed (30 November 2022) | Viewed by 22917

Special Issue Editors


E-Mail Website
Guest Editor
Department of Psychology and the Gonda Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel
Interests: endocannabinoids; biological psychiatry; epigenetics; developmental psychobiology; behavioral neuroscience; psychoneuroendocrinology; animal models of obesity, binge eating, depression, infant–mother attachment, emotion-regulation
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Psychology and the Gonda Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel
Interests: endocannabinoids; early development; psychopathology; mental health; psychoneuroendocrinology; epigenetics; stress exposure; randomized controlled clinical trials
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

This Special Issue is the continuation of our previous special issue "Endocannabinoids, Cannabinoids and Psychiatry: Biological Mechanisms".

During the past decade, scientific knowledge on the role and contributions of endocannabinoids and cannabinoids to the optimal functioning of the human brain has advanced considerably. Currently, the endocannabinoid system is regarded, based on recent scientific findings, as a modulatory system of brain connectivity and top-down bottom-up structural pathways’ protection against extreme inhibitory or extreme excitatory cortical and sub-cortical conditions. Such extreme conditions have been suggested to underlie psychopathology. Additionally, the stabilizing effect of cannabinoids in cases of mood disorders, anxiety, depression and schizophrenia, social phobia, personality disorders, bipolar disorders, and post-traumatic stress disorder has been shown in recent years by various researchers including those using animal models. Research on the role of endocannabinoids in the development of psychiatric disorders, their course and remission has also recently advanced. The field of clinical trials on cannabinoid treatment in psychiatry is just emerging. This timely issue will be among the first to publish novel data on endocannabinoids and cannabinoids in various psychiatric disorders. This is a call for papers on underlying biological, including molecular and neural, mechanisms of endocannabinoid and cannabinoid functions in psychiatric disorders based on animal models, in vitro and ex vivo, models and human clinical trials. Data on age and gender, as well as early development and life span, considerations of endocannabinoids and cannabinoids in psychiatric disorders are also encouraged. Epigenetic and genetic vulnerabilities interacting with the environmental impact related to the endocannabinoid system in psychopathology are also welcome. In this context, magnetic resonance and other imaging studies and studies on crosstalk with other neurotransmitter systems are among the top interests of this issue. The vulnerability of the endocannabinoid system under stressful conditions will also be a focus of this issue, including interactions of this system with stress exposure. Risk factors for the development of psychiatric disorders related to the endocannabinoid system and identified within the fields of brain research, epigenetic, genetic, and molecular research are important components to be included in this issue. Cutting-edge reviews are also invited. We note that non-randomized clinical research and survey studies are not suitable for IJMS.

Prof. Aron Weller
Dr. Sari Goldstein Ferber
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • endocannabinoids
  • cannabinoids
  • psychiatry
  • neural mechanisms
  • molecular mechanisms
  • epigenetics
  • imaging
  • psychopathology
  • stress

Related Special Issue

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

4 pages, 200 KiB  
Editorial
Diverse Underlying Mechanisms and Sex Differences Found in Translational Models of Cannabinoids Use: Towards Validation in Human Studies
by Sari Goldstein Ferber and Aron Weller
Int. J. Mol. Sci. 2023, 24(23), 16586; https://doi.org/10.3390/ijms242316586 - 22 Nov 2023
Viewed by 526
Abstract
This Special Issue represents a continuation of our previous Special Issue entitled “Endocannabinoids, Cannabinoids and Psychiatry: Biological Mechanisms” [...] Full article

Research

Jump to: Editorial, Review

19 pages, 2779 KiB  
Article
Cannabinoid Receptor 2 Blockade Prevents Anti-Depressive-like Effect of Cannabidiol Acid Methyl Ester in Female WKY Rats
by Danielle Hen-Shoval, Lital Moshe, Talia Indig-Naimer, Raphael Mechoulam, Gal Shoval, Gil Zalsman, Natalya M. Kogan and Aron Weller
Int. J. Mol. Sci. 2023, 24(4), 3828; https://doi.org/10.3390/ijms24043828 - 14 Feb 2023
Cited by 4 | Viewed by 2080
Abstract
The pathophysiology of major depressive disorder (MDD) is diverse and multi-factorial, yet treatment strategies remain limited. While women are twice as likely to develop the disorder as men, many animal model studies of antidepressant response rely solely on male subjects. The endocannabinoid system [...] Read more.
The pathophysiology of major depressive disorder (MDD) is diverse and multi-factorial, yet treatment strategies remain limited. While women are twice as likely to develop the disorder as men, many animal model studies of antidepressant response rely solely on male subjects. The endocannabinoid system has been linked to depression in clinical and pre-clinical studies. Cannabidiolic Acid-Methyl Ester (CBDA-ME, EPM-301) demonstrated anti-depressive-like effects in male rats. Here, we explored acute effects of CBDA-ME and some possible mediating mechanisms, using a depressive-like genetic animal model, the Wistar–Kyoto (WKY) rat. In Experiment 1, Female WKY rats underwent the Forced swim test (FST) following acute CBDA-ME oral ingestion (1/5/10 mg/kg). In Experiment 2, Male and female WKY rats underwent the FST after injection of CB1 (AM-251) and CB2 (AM-630) receptor antagonists 30 min before acute CBDA-ME ingestion (1 mg/kg, males; 5 mg/kg, females). Serum levels of Brain-Derived Neurotrophic Factor (BDNF), numerous endocannabinoids and hippocampal Fatty Acid Amide Hydrolase (FAAH) levels were assessed. Results indicate that females required higher doses of CBDA-ME (5 and 10 mg/kg) to induce an anti-depressive-like effect in the FST. AM-630 blocked the antidepressant-like effect in females, but not in males. The effect of CBDA-ME in females was accompanied by elevated serum BDNF and some endocannabinoids and low hippocampal expression of FAAH. This study shows a sexually diverse behavioral anti-depressive response to CBDA-ME and possible underlying mechanisms in females, supporting its potential use for treating MDD and related disorders. Full article
Show Figures

Figure 1

23 pages, 1307 KiB  
Article
FAAH Inhibition Restores Early Life Stress-Induced Alterations in PFC microRNAs Associated with Depressive-Like Behavior in Male and Female Rats
by Anna Portugalov, Hiba Zaidan, Inna Gaisler-Salomon, Cecilia J. Hillard and Irit Akirav
Int. J. Mol. Sci. 2022, 23(24), 16101; https://doi.org/10.3390/ijms232416101 - 17 Dec 2022
Cited by 7 | Viewed by 1505
Abstract
Early life stress (ELS) increases predisposition to depression. We compared the effects of treatment with the fatty acid amide hydrolase (FAAH) inhibitor URB597, and the selective serotonin reuptake inhibitor paroxetine, on ELS-induced depressive-like behavior and the expression of microRNAs (miRs) associated with depression [...] Read more.
Early life stress (ELS) increases predisposition to depression. We compared the effects of treatment with the fatty acid amide hydrolase (FAAH) inhibitor URB597, and the selective serotonin reuptake inhibitor paroxetine, on ELS-induced depressive-like behavior and the expression of microRNAs (miRs) associated with depression in the medial prefrontal cortex (mPFC), hippocampal CA1 area, lateral habenula and dorsal raphe in rats. We also examined the mRNA expression of serotonergic (htr1a and slc6a4) and endocannabinoid (cnr1, cnr2 and faah) targets in the mPFC following ELS and pharmacological treatment. Adult males and females exposed to the ‘Limited Bedding and Nesting’ ELS paradigm demonstrated a depressive-like phenotype and late-adolescence URB597 treatment, but not paroxetine, reversed this phenotype. In the mPFC, ELS downregulated miR-16 in males and miR-135a in females and URB597 treatment restored this effect. In ELS females, the increase in cnr2 and decrease in faah mRNAs in the mPFC were reversed by URB597 treatment. We show for the first time that URB597 reversed ELS-induced mPFC downregulation in specific miRs and stress-related behaviors, suggesting a novel mechanism for the beneficial effects of FAAH inhibition. The differential effects of ELS and URB597 on males and females highlight the importance of developing sex-specific treatment approaches. Full article
Show Figures

Figure 1

14 pages, 3270 KiB  
Article
Cannabidiol Prevents Spontaneous Fear Recovery after Extinction and Ameliorates Stress-Induced Extinction Resistance
by Eleni P. Papagianni, William G. Warren, Helen J. Cassaday and Carl W. Stevenson
Int. J. Mol. Sci. 2022, 23(16), 9333; https://doi.org/10.3390/ijms23169333 - 19 Aug 2022
Cited by 2 | Viewed by 2389
Abstract
Cannabidiol, the main non-psychotropic constituent of cannabis, has potential as a treatment for anxiety-related disorders since it reduces learned fear expression and enhances fear extinction. The return of fear over time after successful extinction and stress-induced extinction resistance are potential barriers to the [...] Read more.
Cannabidiol, the main non-psychotropic constituent of cannabis, has potential as a treatment for anxiety-related disorders since it reduces learned fear expression and enhances fear extinction. The return of fear over time after successful extinction and stress-induced extinction resistance are potential barriers to the treatment of these disorders with extinction-based psychological therapy. In two experiments using rats subjected to auditory fear conditioning, we determined the effects of systemic cannabidiol treatment on (1) delayed extinction and later spontaneous fear recovery, and (2) extinction resistance caused by immediate extinction (the immediate extinction deficit (IED)). In Experiment 1, cannabidiol was given before delayed extinction occurring 24 h after conditioning, with extinction recall and spontaneous fear recovery tested drug-free 1 and 21 days after extinction, respectively. We found that cannabidiol had no effect on extinction recall but it prevented spontaneous fear recovery. In Experiment 2, the IED procedure was first validated, with immediate extinction occurring 30 min after conditioning. We confirmed that immediate extinction impaired extinction recall, compared to delayed extinction. Next, cannabidiol was given before immediate or no extinction, with extinction recall tested drug-free the next day. We found that cannabidiol rescued the IED, which did not involve effects on fear memory consolidation. In summary, cannabidiol prevented spontaneous fear recovery after delayed extinction and ameliorated extinction resistance caused by immediate extinction. Although the pharmacological mechanisms underlying these effects remain to be determined, our results add to evidence indicating that cannabidiol might prove useful as an adjunct for potentiating the psychological treatment of anxiety-related disorders. Full article
Show Figures

Figure 1

22 pages, 2513 KiB  
Article
Cannabinoid-Induced Conditioned Place Preference, Intravenous Self-Administration, and Behavioral Stimulation Influenced by Ghrelin Receptor Antagonism in Rats
by Chrysostomos Charalambous, Tereza Havlickova, Marek Lapka, Nina Puskina, Romana Šlamberová, Martin Kuchar and Magdalena Sustkova-Fiserova
Int. J. Mol. Sci. 2021, 22(5), 2397; https://doi.org/10.3390/ijms22052397 - 27 Feb 2021
Cited by 10 | Viewed by 2592
Abstract
Cannabis/cannabinoids are widely used for recreational and therapy purposes, but their risks are largely disregarded. However, cannabinoid-associated use disorders and dependence are alarmingly increasing and an effective treatment is lacking. Recently, the growth hormone secretagogue receptor (GHSR1A) antagonism was proposed as a promising [...] Read more.
Cannabis/cannabinoids are widely used for recreational and therapy purposes, but their risks are largely disregarded. However, cannabinoid-associated use disorders and dependence are alarmingly increasing and an effective treatment is lacking. Recently, the growth hormone secretagogue receptor (GHSR1A) antagonism was proposed as a promising mechanism for drug addiction therapy. However, the role of GHS-R1A and its endogenous ligand ghrelin in cannabinoid abuse remains unclear. Therefore, the aim of our study was to investigate whether the GHS-R1A antagonist JMV2959 could reduce the tetrahydrocannabinol (THC)-induced conditioned place preference (CPP) and behavioral stimulation, the WIN55,212-2 intravenous self-administration (IVSA), and the tendency to relapse. Following an ongoing WIN55,212-2 self-administration, JMV2959 3 mg/kg was administered intraperitoneally 20 min before three consequent daily 120-min IVSA sessions under a fixed ratio FR1, which significantly reduced the number of the active lever-pressing, the number of infusions, and the cannabinoid intake. Pretreatment with JMV2959 suggested reduction of the WIN55,212-2-seeking/relapse-like behavior tested in rats on the twelfth day of the forced abstinence period. On the contrary, pretreatment with ghrelin significantly increased the cannabinoid IVSA as well as enhanced the relapse-like behavior. Co-administration of ghrelin with JMV2959 abolished/reduced the significant efficacy of the GHS-R1A antagonist in the cannabinoid IVSA. Pretreatment with JMV2959 significantly and dose-dependently reduced the manifestation of THC-induced CPP. The THC-CPP development was reduced after the simultaneous administration of JMV2959 with THC during conditioning. JMV2959 also significantly reduced the THC-induced behavioral stimulation in the LABORAS cage. Our findings suggest that GHS-R1A importantly participates in the rewarding/reinforcing effects of cannabinoids. Full article
Show Figures

Figure 1

Review

Jump to: Editorial, Research

25 pages, 440 KiB  
Review
Modulation of Endocannabinoid System Components in Depression: Pre-Clinical and Clinical Evidence
by Uri Bright and Irit Akirav
Int. J. Mol. Sci. 2022, 23(10), 5526; https://doi.org/10.3390/ijms23105526 - 15 May 2022
Cited by 11 | Viewed by 3401
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 [...] Read more.
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. Full article
19 pages, 963 KiB  
Review
Pro-Inflammatory Cytokines: Potential Links between the Endocannabinoid System and the Kynurenine Pathway in Depression
by Ferenc Zádor, Sâmia Joca, Gábor Nagy-Grócz, Szabolcs Dvorácskó, Edina Szűcs, Csaba Tömböly, Sándor Benyhe and László Vécsei
Int. J. Mol. Sci. 2021, 22(11), 5903; https://doi.org/10.3390/ijms22115903 - 31 May 2021
Cited by 27 | Viewed by 4561
Abstract
Substance use/abuse is one of the main causes of depressive symptoms. Cannabis and synthetic cannabinoids in particular gained significant popularity in the past years. There is an increasing amount of clinical data associating such compounds with the inflammatory component of depression, indicated by [...] Read more.
Substance use/abuse is one of the main causes of depressive symptoms. Cannabis and synthetic cannabinoids in particular gained significant popularity in the past years. There is an increasing amount of clinical data associating such compounds with the inflammatory component of depression, indicated by the up-regulation of pro-inflammatory cytokines. Pro-inflammatory cytokines are also well-known to regulate the enzymes of the kynurenine pathway (KP), which is responsible for metabolizing tryptophan, a precursor in serotonin synthesis. Enhanced pro-inflammatory cytokine levels may over-activate the KP, leading to tryptophan depletion and reduced serotonin levels, which can subsequently precipitate depressive symptoms. Therefore, such mechanism might represent a possible link between the endocannabinoid system (ECS) and the KP in depression, via the inflammatory and dysregulated serotonergic component of the disorder. This review will summarize the data regarding those natural and synthetic cannabinoids that increase pro-inflammatory cytokines. Furthermore, the data on such cytokines associated with KP activation will be further reviewed accordingly. The interaction of the ECS and the KP has been postulated and demonstrated in some studies previously. This review will further contribute to this yet less explored connection and propose the KP to be the missing link between cannabinoid-induced inflammation and depressive symptoms. Full article
Show Figures

Graphical abstract

20 pages, 2765 KiB  
Review
The (Poly)Pharmacology of Cannabidiol in Neurological and Neuropsychiatric Disorders: Molecular Mechanisms and Targets
by Rosa Maria Vitale, Fabio Arturo Iannotti and Pietro Amodeo
Int. J. Mol. Sci. 2021, 22(9), 4876; https://doi.org/10.3390/ijms22094876 - 5 May 2021
Cited by 36 | Viewed by 4831
Abstract
Cannabidiol (CBD), the major nonpsychoactive Cannabis constituent, has been proposed for the treatment of a wide panel of neurological and neuropsychiatric disorders, including anxiety, schizophrenia, epilepsy and drug addiction due to the ability of its versatile scaffold to interact with diverse molecular targets [...] Read more.
Cannabidiol (CBD), the major nonpsychoactive Cannabis constituent, has been proposed for the treatment of a wide panel of neurological and neuropsychiatric disorders, including anxiety, schizophrenia, epilepsy and drug addiction due to the ability of its versatile scaffold to interact with diverse molecular targets that are not restricted to the endocannabinoid system. Albeit the molecular mechanisms responsible for the therapeutic effects of CBD have yet to be fully elucidated, many efforts have been devoted in the last decades to shed light on its complex pharmacological profile. In particular, an ever-increasing number of molecular targets linked to those disorders have been identified for this phytocannabinoid, along with the modulatory effects of CBD on their cascade signaling. In this view, here we will try to provide a comprehensive and up-to-date overview of the molecular basis underlying the therapeutic effects of CBD involved in the treatment of neurological and neuropsychiatric disorders. Full article
Show Figures

Figure 1

Back to TopTop