1
|
Maehashi S, Arora K, Fisher AL, Schweitzer DR, Akefe IO. Neurolipidomic insights into anxiety disorders: Uncovering lipid dynamics for potential therapeutic advances. Neurosci Biobehav Rev 2024; 163:105741. [PMID: 38838875 DOI: 10.1016/j.neubiorev.2024.105741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
Anxiety disorders constitute a spectrum of psychological conditions affecting millions of individuals worldwide, imposing a significant health burden. Historically, the development of anxiolytic medications has been largely focused on neurotransmitter function and modulation. However, in recent years, neurolipids emerged as a prime target for understanding psychiatric pathogenesis and developing novel medications. Neurolipids influence various neural activities such as neurotransmission and cellular functioning, as well as maintaining cell membrane integrity. Therefore, this review aims to elucidate the alterations in neurolipids associated with an anxious mental state and explore their potential as targets of novel anxiolytic medications. Existing evidence tentatively associates dysregulated neurolipid levels with the etiopathology of anxiety disorders. Notably, preclinical investigations suggest that several neurolipids, including endocannabinoids and polyunsaturated fatty acids, may hold promise as potential pharmacological targets. Overall, the current literature tentatively suggests the involvement of lipids in the pathogenesis of anxiety disorders, hinting at potential prospects for future pharmacological interventions.
Collapse
Affiliation(s)
- Saki Maehashi
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
| | - Kabir Arora
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andre Lara Fisher
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | | | - Isaac Oluwatobi Akefe
- Academy for Medical Education, The University of Queensland, Herston, QLD 4006, Australia.
| |
Collapse
|
2
|
Zhao H, Liu Y, Cai N, Liao X, Tang L, Wang Y. Endocannabinoid Hydrolase Inhibitors: Potential Novel Anxiolytic Drugs. Drug Des Devel Ther 2024; 18:2143-2167. [PMID: 38882045 PMCID: PMC11179644 DOI: 10.2147/dddt.s462785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024] Open
Abstract
Over the past decade, the idea of targeting the endocannabinoid system to treat anxiety disorders has received increasing attention. Previous studies focused more on developing cannabinoid receptor agonists or supplementing exogenous cannabinoids, which are prone to various adverse effects due to their strong pharmacological activity and poor receptor selectivity, limiting their application in clinical research. Endocannabinoid hydrolase inhibitors are considered to be the most promising development strategies for the treatment of anxiety disorders. More recent efforts have emphasized that inhibition of two major endogenous cannabinoid hydrolases, monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH), indirectly activates cannabinoid receptors by increasing endogenous cannabinoid levels in the synaptic gap, circumventing receptor desensitization resulting from direct enhancement of endogenous cannabinoid signaling. In this review, we comprehensively summarize the anxiolytic effects of MAGL and FAAH inhibitors and their potential pharmacological mechanisms, highlight reported novel inhibitors or natural products, and provide an outlook on future directions in this field.
Collapse
Affiliation(s)
- Hongqing Zhao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| | - Yang Liu
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| | - Na Cai
- Outpatient Department, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Xiaolin Liao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| | - Lin Tang
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
- Department of Pharmacy, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Yuhong Wang
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
3
|
Fusse EJ, Scarante FF, Vicente MA, Marrubia MM, Turcato F, Scomparin DS, Ribeiro MA, Figueiredo MJ, Brigante TAV, Guimarães FS, Campos AC. Anxiogenic doses of rapamycin prevent URB597-induced anti-stress effects in socially defeated mice. Neurosci Lett 2024; 818:137519. [PMID: 37852528 DOI: 10.1016/j.neulet.2023.137519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Repeated exposure to psychosocial stress modulates the endocannabinoid system, particularly anandamide (AEA) signaling in brain regions associated with emotional distress. The mTOR protein regulates various neuroplastic processes in the brain disrupted by stress, including adult hippocampal neurogenesis. This kinase has been implicated in multiple effects of cannabinoid drugs and the anti-stress behavioral effects of psychoactive drugs. Therefore, our hypothesis is that enhancing AEA signaling via pharmacological inhibition of the fatty acid amide hydrolase (FAAH) enzyme induces an anti-stress behavioral effect through an mTOR-dependent mechanism. To test this hypothesis, male C57Bl6 mice were exposed to social defeat stress (SDS) for 7 days and received daily treatment with either vehicle or different doses of the FAAH inhibitor, URB597 (0.1; 0.3; 1 mg/Kg), alone or combined with rapamycin. The results suggested that URB597 induced an inverted U-shaped dose-response curve in mice subjected to SDS (with the intermediate dose of 0.3 mg/kg being anxiolytic, and the higher tested dose of 1 mg/Kg being anxiogenic). In a second independent experiment, rapamycin treatment induced an anxiogenic-like response in control mice. However, in the presence of rapamycin, the anxiolytic dose of URB597 treatment failed to reduce stress-induced anxiety behaviors in mice. SDS exposure altered the hippocampal expression of the mTOR scaffold protein Raptor. Furthermore, the anxiogenic dose of URB597 decreased the absolute number of migrating doublecortin (DCX)-positive cells in the dentate gyrus, suggesting an anti-anxiety effect independent of newly generated/immature neurons. Therefore, our results indicate that in mice exposed to repeated psychosocial stress, URB597 fails to counteract the anxiogenic-like response induced by the pharmacological dampening of mTOR signaling.
Collapse
Affiliation(s)
- Eduardo J Fusse
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Franciele F Scarante
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Maria A Vicente
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Mariana M Marrubia
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Flávia Turcato
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, USA
| | - Davi S Scomparin
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Melissa A Ribeiro
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Maria J Figueiredo
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Tamires A V Brigante
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Alline C Campos
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil.
| |
Collapse
|
4
|
Voicu V, Brehar FM, Toader C, Covache-Busuioc RA, Corlatescu AD, Bordeianu A, Costin HP, Bratu BG, Glavan LA, Ciurea AV. Cannabinoids in Medicine: A Multifaceted Exploration of Types, Therapeutic Applications, and Emerging Opportunities in Neurodegenerative Diseases and Cancer Therapy. Biomolecules 2023; 13:1388. [PMID: 37759788 PMCID: PMC10526757 DOI: 10.3390/biom13091388] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
In this review article, we embark on a thorough exploration of cannabinoids, compounds that have garnered considerable attention for their potential therapeutic applications. Initially, this article delves into the fundamental background of cannabinoids, emphasizing the role of endogenous cannabinoids in the human body and outlining their significance in studying neurodegenerative diseases and cancer. Building on this foundation, this article categorizes cannabinoids into three main types: phytocannabinoids (plant-derived cannabinoids), endocannabinoids (naturally occurring in the body), and synthetic cannabinoids (laboratory-produced cannabinoids). The intricate mechanisms through which these compounds interact with cannabinoid receptors and signaling pathways are elucidated. A comprehensive overview of cannabinoid pharmacology follows, highlighting their absorption, distribution, metabolism, and excretion, as well as their pharmacokinetic and pharmacodynamic properties. Special emphasis is placed on the role of cannabinoids in neurodegenerative diseases, showcasing their potential benefits in conditions such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. The potential antitumor properties of cannabinoids are also investigated, exploring their potential therapeutic applications in cancer treatment and the mechanisms underlying their anticancer effects. Clinical aspects are thoroughly discussed, from the viability of cannabinoids as therapeutic agents to current clinical trials, safety considerations, and the adverse effects observed. This review culminates in a discussion of promising future research avenues and the broader implications for cannabinoid-based therapies, concluding with a reflection on the immense potential of cannabinoids in modern medicine.
Collapse
Affiliation(s)
- Victor Voicu
- Pharmacology, Toxicology and Clinical Psychopharmacology, “Carol Davila” University of Medicine and Pharmacy in Bucharest, 020021 Bucharest, Romania;
- Medical Section within the Romanian Academy, 010071 Bucharest, Romania
| | - Felix-Mircea Brehar
- Neurosurgery Department, Emergency Clinical Hospital Bagdasar-Arseni, 041915 Bucharest, Romania
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Antonio Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Andrei Bordeianu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Horia Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Luca-Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
5
|
Fang G, Wang Y. Hippocampal CB1 receptor mediates antidepressant-like effect of synthetic cannabinoid-HU210 in acute despair reaction model in mice. Neurosci Lett 2023; 792:136953. [PMID: 36372093 DOI: 10.1016/j.neulet.2022.136953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/23/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022]
Abstract
Growing evidence suggests that stress may contribute to the pathophysiology of depression. The alleviation of depressive symptoms is one of the most attractive medical applications of cannabis. Here, we investigated the antidepressant-like actions of synthetic cannabinoid-HU210 in acute despair response and explored the possible underlying mechanisms. Acute stress, induced by forced-swimming, induced depression-like behavior in the sucrose preference test (SPT). HU-210 (50 μg/kg) displayed anti-depressant like effect in the forced swim test in naïve mice and decreased depression-like behavior in the SPT, induced by forced swim stress. Pretreatment with AM251, an inhibitor of CB1R or inhibition of long-term depression (LTD) at hippocampal CA3-CA1 synapses by Tat-GluR2 attenuated the antidepressant like action of HU-210. These results indicate that HU210 produces antidepressant-like effects in acute stress and its underlying mechanism may be related to CB1R activation and hence hippocampal LTD production invivo. Synthetic cannabis or cannabis-related drugs may be used as an early intervention after acute stress exposure to prevent or at least reduce depression-like behaviors.
Collapse
Affiliation(s)
- Guoxiang Fang
- Department of Emergency, Third Hospital of Xi'an, 10(#) Fengcheng Three Road, Xi'an, Shaanxi 710018, China
| | - Ying Wang
- Department of Psychiatry, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, China.
| |
Collapse
|
6
|
Gilio L, Fresegna D, Gentile A, Guadalupi L, Sanna K, De Vito F, Balletta S, Caioli S, Rizzo FR, Musella A, Iezzi E, Moscatelli A, Galifi G, Fantozzi R, Bellantonio P, Furlan R, Finardi A, Vanni V, Dolcetti E, Bruno A, Buttari F, Mandolesi G, Centonze D, Stampanoni Bassi M. Preventive exercise attenuates IL-2-driven mood disorders in multiple sclerosis. Neurobiol Dis 2022; 172:105817. [PMID: 35835361 DOI: 10.1016/j.nbd.2022.105817] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 10/17/2022] Open
Abstract
BACKGROUND Elevated levels of specific proinflammatory molecules in the cerebrospinal fluid (CSF) have been associated with disability progression, enhanced neurodegeneration and higher incidence of mood disorders in people with multiple sclerosis (MS). Studies in animal models of MS suggest that preventive exercise may play an immunomodulatory activity, with beneficial effects on both motor deficits and behavioral alterations. Here we explored the impact of lifestyle physical activity on clinical presentation and associated central inflammation in a large group of newly diagnosed patients with MS. Furthermore, we addressed the causal link between exercise-mediated immunomodulation and mood symptoms in the animal setting. METHODS A cross-sectional study was conducted on 235 relapsing-remitting MS patients at the time of the diagnosis. Patients were divided into 3 groups ("sedentary", "lifestyle physical activity" and "exercise") according to the level of physical activity in the six months preceding the evaluation. Patients underwent clinical, neuropsychological and psychiatric evaluation, magnetic resonance imaging and lumbar puncture for diagnostic purposes. The CSF levels of proinflammatory and anti-inflammatory cytokines were analyzed and compared with a group of 80 individuals with non-inflammatory and non-degenerative diseases. Behavioral and electrophysiological studies were carried out in control mice receiving intracerebral injection of IL-2 or vehicle. Behavior was also assessed in mice with experimental autoimmune encephalomyelitis (EAE), animal model of MS, reared in standard (sedentary group) or running wheel-equipped (exercise group) cages. RESULTS In exercising MS patients, depression and anxiety were reduced compared to sedentary patients. The CSF levels of the interleukin-2 and 6 (IL-2, IL-6) were increased in MS patients compared with control individuals. In MS subjects exercise was associated with normalized CSF levels of IL-2. In EAE mice exercise started before disease onset reduced both behavioral alterations and striatal IL-2 expression. Notably, a causal role of IL-2 in mood disorders was shown. IL-2 administration in control healthy mice induced anxious- and depressive-like behaviors and impaired type-1 cannabinoid (CB1) receptor-mediated neurotransmission at GABAergic synapses, mimicking EAE-induced synaptic dysfunction. CONCLUSIONS Our results indicate an immunomodulatory effect of exercise in MS patients, associated with reduced CSF expression of IL-2, which might result in reduced mood disorders. These data suggest that exercise in the early stages may act as a disease-modifying therapy in MS although further longitudinal studies are needed to clarify this issue.
Collapse
Affiliation(s)
- Luana Gilio
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
| | | | - Livia Guadalupi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy; Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Krizia Sanna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy; Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | | - Sara Balletta
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy; Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Silvia Caioli
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | | | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy; Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
| | - Ennio Iezzi
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Alessandro Moscatelli
- Department of Systems Medicine, Tor Vergata University, Rome, Italy; Laboratory of Neuromotor Physiology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | | | | | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
| | | | - Antonio Bruno
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy; Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
| | - Diego Centonze
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy; Department of Systems Medicine, Tor Vergata University, Rome, Italy.
| | | |
Collapse
|
7
|
Enhancing Endocannabinoid Control of Stress with Cannabidiol. J Clin Med 2021; 10:jcm10245852. [PMID: 34945148 PMCID: PMC8704602 DOI: 10.3390/jcm10245852] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 02/08/2023] Open
Abstract
The stress response is a well-defined physiological function activated frequently by life events. However, sometimes the stress response can be inappropriate, excessive, or prolonged; in which case, it can hinder rather than help in coping with the stressor, impair normal functioning, and increase the risk of somatic and mental health disorders. There is a need for a more effective and safe pharmacological treatment that can dampen maladaptive stress responses. The endocannabinoid system is one of the main regulators of the stress response. A basal endocannabinoid tone inhibits the stress response, modulation of this tone permits/curtails an active stress response, and chronic deficiency in the endocannabinoid tone is associated with the pathological complications of chronic stress. Cannabidiol is a safe exogenous cannabinoid enhancer of the endocannabinoid system that could be a useful treatment for stress. There have been seven double-blind placebo controlled clinical trials of CBD for stress on a combined total of 232 participants and one partially controlled study on 120 participants. All showed that CBD was effective in significantly reducing the stress response and was non-inferior to pharmaceutical comparators, when included. The clinical trial results are supported by the established mechanisms of action of CBD (including increased N-arachidonylethanolamine levels) and extensive real-world and preclinical evidence of the effectiveness of CBD for treating stress.
Collapse
|
8
|
Pietropaolo S, Marsicano G. The role of the endocannabinoid system as a therapeutic target for autism spectrum disorder: Lessons from behavioral studies on mouse models. Neurosci Biobehav Rev 2021; 132:664-678. [PMID: 34813825 DOI: 10.1016/j.neubiorev.2021.11.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 11/02/2021] [Accepted: 11/19/2021] [Indexed: 12/17/2022]
Abstract
Recent years have seen an impressive amount of research devoted to understanding the etiopathology of Autism Spectrum Disorder (ASD) and developing therapies for this syndrome. Because of the lack of biomarkers of ASD, this work has been largely based on the behavioral characterization of rodent models, based on a multitude of genetic and environmental manipulations. Here we highlight how the endocannabinoid system (ECS) has recently emerged within this context of mouse behavioral studies as an etiopathological factor in ASD and a valid potential therapeutic target. We summarize the most recent results showing alterations of the ECS in rodent models of ASD, and demonstrating ASD-like behaviors in mice with altered ECS, induced either by genetic or pharmacological manipulations. We also give a critical overview of the most relevant advances in designing treatments and novel mouse models for ASD targeting the ECS, highlighting the relevance of thorough and innovative behavioral approaches to investigate the mechanisms acting underneath the complex features of ASD.
Collapse
Affiliation(s)
| | - Giovanni Marsicano
- INSERM, U1215 NeuroCentre Magendie, 146 rue Léo Saignat, 33077, Bordeaux Cedex, France
| |
Collapse
|
9
|
Petrie GN, Nastase AS, Aukema RJ, Hill MN. Endocannabinoids, cannabinoids and the regulation of anxiety. Neuropharmacology 2021; 195:108626. [PMID: 34116110 DOI: 10.1016/j.neuropharm.2021.108626] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/11/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022]
Abstract
Cannabis has been used for hundreds of years, with its ability to dampen feelings of anxiety often reported as a primary reason for use. Only recently has the specific role cannabinoids play in anxiety been thoroughly investigated. Here we discuss the body of evidence describing how endocannabinoids and exogenous cannabinoids are capable of regulating the generation and termination of anxiety states. Disruption of the endogenous cannabinoid (eCB) system following genetic manipulation, pharmacological intervention or stress exposure reliably leads to the generation of an anxiety state. On the other hand, upregulation of eCB signaling is capable of alleviating anxiety-like behaviors in multiple paradigms. When considering exogenous cannabinoid administration, cannabinoid receptor 1 (CB1) agonists have a biphasic, dose-dependent effect on anxiety such that low doses are anxiolytic while high doses are anxiogenic, a phenomenon that is evident in both rodent models and humans. Translational studies investigating a loss of function mutation in the gene for fatty acid amide hydrolase, the enzyme responsible for metabolizing AEA, have also shown that AEA signaling regulates anxiety in humans. Taken together, evidence reviewed here has outlined a convincing argument for cannabinoids being powerful regulators of both the manifestation and amelioration of anxiety symptoms, and highlights the therapeutic potential of targeting the eCB system for the development of novel classes of anxiolytics. This article is part of the special issue on 'Cannabinoids'.
Collapse
Affiliation(s)
- Gavin N Petrie
- Hotchkiss Brain Institute and the Mathison Centre for Mental Health Education and Research, Departments of Cell Biology and Anatomy & Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Andrei S Nastase
- Hotchkiss Brain Institute and the Mathison Centre for Mental Health Education and Research, Departments of Cell Biology and Anatomy & Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Robert J Aukema
- Hotchkiss Brain Institute and the Mathison Centre for Mental Health Education and Research, Departments of Cell Biology and Anatomy & Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute and the Mathison Centre for Mental Health Education and Research, Departments of Cell Biology and Anatomy & Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
10
|
Wilkerson JL, Bilbrey JA, Felix JS, Makriyannis A, McMahon LR. Untapped endocannabinoid pharmacological targets: Pipe dream or pipeline? Pharmacol Biochem Behav 2021; 206:173192. [PMID: 33932409 DOI: 10.1016/j.pbb.2021.173192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
It has been established that the endogenous cannabinoid (endocannabinoid) system plays key modulatory roles in a wide variety of pathological conditions. The endocannabinoid system comprises both cannabinoid receptors, their endogenous ligands including 2-arachidonoylglycerol (2-AG), N-arachidonylethanolamine (anandamide, AEA), and enzymes that regulate the synthesis and degradation of endogenous ligands which include diacylglycerol lipase alpha (DAGL-α), diacylglycerol lipase beta (DAGL-β), fatty acid amide hydrolase (FAAH), monoacylglycerol lipase (MAGL), α/β hydrolase domain 6 (ABHD6). As the endocannabinoid system exerts considerable involvement in the regulation of homeostasis and disease, much effort has been made towards understanding endocannabinoid-related mechanisms of action at cellular, physiological, and pathological levels as well as harnessing the various components of the endocannabinoid system to produce novel therapeutics. However, drug discovery efforts within the cannabinoid field have been slower than anticipated to reach satisfactory clinical endpoints and raises an important question into the validity of developing novel ligands that therapeutically target the endocannabinoid system. To answer this, we will first examine evidence that supports the existence of an endocannabinoid system role within inflammatory diseases, neurodegeneration, pain, substance use disorders, mood disorders, as well as metabolic diseases. Next, this review will discuss recent clinical studies, within the last 5 years, of cannabinoid compounds in context to these diseases. We will also address some of the challenges and considerations within the cannabinoid field that may be important in the advancement of therapeutics into the clinic.
Collapse
Affiliation(s)
- Jenny L Wilkerson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| | - Joshua A Bilbrey
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Jasmine S Felix
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; Departments of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Lance R McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
11
|
Restoring glutamate homeostasis in the nucleus accumbens via endocannabinoid-mimetic drug prevents relapse to cocaine seeking behavior in rats. Neuropsychopharmacology 2021; 46:970-981. [PMID: 33514875 PMCID: PMC8115336 DOI: 10.1038/s41386-021-00955-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 12/13/2020] [Accepted: 12/29/2020] [Indexed: 12/23/2022]
Abstract
Impaired glutamate homeostasis is a key characteristic of the neurobiology of drug addiction in rodent models and contributes to the vulnerability to relapse to drug seeking. Although disrupted astrocytic and presynaptic regulation of glutamate release has been considered to constitute with impaired glutamate homeostasis in rodent model of drug relapse, the involvement of endocannabinoids (eCBs) in this neurobiological process has remained largely unknown. Here, using cocaine self-administration in rats, we investigated the role of endocannabinoids in impaired glutamate homeostasis in the core of nucleus accumbens (NAcore), which was indicated by augmentation of spontaneous synaptic glutamate release, downregulation of metabotropic glutamate receptor 2/3 (mGluR2/3), and mGluR5-mediated astrocytic glutamate release. We found that the endocannabinoid, anandamide (AEA), rather than 2-arachidonoylglycerol elicited glutamate release through presynaptic transient receptor potential vanilloid 1 (TRPV1) and astrocytic cannabinoid type-1 receptors (CB1Rs) in the NAcore of saline-yoked rats. In rats with a history of cocaine self-administration and extinction training, AEA failed to alter synaptic glutamate release in the NAcore, whereas CB1R-mediated astrocytic glutamate release by AEA remained functional. In order to induce increased astrocytic glutamate release via exogenous AEA, (R)-methanandamide (methAEA, a metabolically stable form of AEA) was chronically infused in the NAcore via osmotic pumps during extinction training. Restoration of mGluR2/3 function and mGluR5-mediated astrocytic glutamate release was observed after chronic methAEA infusion. Additionally, priming or cue-induced reinstatement of cocaine seeking was inhibited in methAEA-infused rats. These results demonstrate that enhancing endocannabinoid signaling is a potential pathway to restore glutamate homeostasis and may represent a promising therapeutic strategy for preventing cocaine relapse.
Collapse
|
12
|
Cannabis use and posttraumatic stress disorder comorbidity: Epidemiology, biology and the potential for novel treatment approaches. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 157:143-193. [PMID: 33648669 DOI: 10.1016/bs.irn.2020.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cannabis use is increasing among some demographics in the United States and is tightly linked to anxiety, trauma, and stress reactivity at the epidemiological and biological level. Stress-coping motives are highly cited reasons for cannabis use. However, with increased cannabis use comes the increased susceptibility for cannabis use disorder (CUD). Indeed, CUD is highly comorbid with posttraumatic stress disorder (PTSD). Importantly, endogenous cannabinoid signaling systems play a key role in the regulation of stress reactivity and anxiety regulation, and preclinical data suggest deficiencies in this signaling system could contribute to the development of stress-related psychopathology. Furthermore, endocannabinoid deficiency states, either pre-existing or induced by trauma exposure, could provide explanatory insights into the high rates of comorbid cannabis use in patients with PTSD. Here we review clinical and preclinical literature related to the cannabis use-PTSD comorbidity, the role of endocannabinoids in the regulation of stress reactivity, and potential therapeutic implications of recent work in this area.
Collapse
|
13
|
Fang G, Wang Y. Transcranial direct current stimulation (tDCS) produce anti-anxiety response in acute stress exposure rats via activation of amygdala CB1R. Behav Brain Res 2020; 400:113050. [PMID: 33279640 DOI: 10.1016/j.bbr.2020.113050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 11/26/2022]
Abstract
Anxiety is one of the most common mental disorders worldwide. Currently, the main anti-anxiety drugs, selective serotonin/noradrenalin reuptake inhibitors (SSRIs/SNRIs), are always associated with delayed onset of action and low therapeutic response rate. Benzodiazepines can produce rapid effects, but their long-term use may result in severe adverse reaction and drug dependence. Transcranial direct current stimulation (tDCS) is one of the noteworthy noninvasive brain stimulation techniques and is expected to be a new choice of anti-anxiety therapy. However, the underlying mechanism remains unclear. In our recent published study, we have observed the important role of endogenous cannabinoid in the pathophysiology and treatment of anxiety. Here we verified the anti-anxiety effects of tDCS in the acute stress exposure rats, and investigated the possible role of amygdala cannabinoid receptor 1 (CB1R) activation in the anti-anxiety response of tDCS. Forced swimming exposure produced anxiety-like behaviors, which can be reversed by tDCS treatment. tDCS increased the time spent in the center without affection of locomotor activity in open field test (OFT) and elevated the number of entries into open arm and time spent in open arm in elevated plus maze test (EPMT). However, Inhibition of CB1R function by AM251 intraperitoneal injection or CB1R knockdown in amygdala produced the negative effects on the anti-anxiety action of tDCS. In conclusion, tDCS may play an anti-anxiety role at least partly via activation of amygdala CB1R, which provides a theoretical basis for the clinical application of tDCS in the treatment of anxiety disorder.
Collapse
Affiliation(s)
- Guoxiang Fang
- Department of Emergency, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710018, PR China
| | - Ying Wang
- Department of Psychiatry, Xi'an International Medical Center Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710100, China.
| |
Collapse
|
14
|
Bedse G, Hill MN, Patel S. 2-Arachidonoylglycerol Modulation of Anxiety and Stress Adaptation: From Grass Roots to Novel Therapeutics. Biol Psychiatry 2020; 88:520-530. [PMID: 32197779 PMCID: PMC7486996 DOI: 10.1016/j.biopsych.2020.01.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/31/2019] [Accepted: 01/18/2020] [Indexed: 01/13/2023]
Abstract
Over the past decade there has been a surge of interest in the development of endocannabinoid-based therapeutic approaches for the treatment of diverse neuropsychiatric conditions. Although initial preclinical and clinical development efforts focused on pharmacological inhibition of fatty acid amide hydrolase to elevate levels of the endocannabinoid anandamide, more recent efforts have focused on inhibition of monoacylglycerol lipase (MAGL) to enhance signaling of the most abundant and efficacious endocannabinoid ligand, 2-arachidonoylglycerol (2-AG). We review the biochemistry and physiology of 2-AG signaling and preclinical evidence supporting a role for this system in the regulation of anxiety-related outcomes and stress adaptation. We review preclinical evidence supporting MAGL inhibition for the treatment of affective, trauma-related, and stress-related disorders; describe the current state of MAGL inhibitor drug development; and discuss biological factors that could affect MAGL inhibitor efficacy. Issues related to the clinical advancement of MAGL inhibitors are also discussed. We are cautiously optimistic, as the field of MAGL inhibitor development transitions from preclinical to clinical and theoretical to practical, that pharmacological 2-AG augmentation could represent a mechanistically novel therapeutic approach for the treatment of affective and stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gaurav Bedse
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Vanderbilt Center for Addiction Research, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mathew N Hill
- Department of Cell Biology, Hotchkiss Brain Institute and Mathison Center for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Anatomy and Psychiatry, Hotchkiss Brain Institute and Mathison Center for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sachin Patel
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Vanderbilt Center for Addiction Research, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
15
|
deRoon-Cassini TA, Stollenwerk TM, Beatka M, Hillard CJ. Meet Your Stress Management Professionals: The Endocannabinoids. Trends Mol Med 2020; 26:953-968. [PMID: 32868170 PMCID: PMC7530069 DOI: 10.1016/j.molmed.2020.07.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/24/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
Abstract
The endocannabinoid signaling system (ECSS) is altered by exposure to stress and mediates and modulates the effects of stress on the brain. Considerable preclinical data support critical roles for the endocannabinoids and their target, the CB1 cannabinoid receptor, in the adaptation of the brain to repeated stress exposure. Chronic stress exposure increases vulnerability to mental illness, so the ECSS has attracted attention as a potential therapeutic target for the prevention and treatment of stress-related psychopathology. We discuss human genetic studies indicating that the ECSS contributes to risk for mental illness in those exposed to severe stress and trauma early in life, and we explore the potential difficulties in pharmacological manipulation of the ECSS.
Collapse
Affiliation(s)
- Terri A deRoon-Cassini
- Neuroscience Research Center, USA; Department of Surgery, Division of Trauma and Acute Care Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Todd M Stollenwerk
- Neuroscience Research Center, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Margaret Beatka
- Neuroscience Research Center, USA; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cecilia J Hillard
- Neuroscience Research Center, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
16
|
Pietropaolo S, Bellocchio L, Bouzón-Arnáiz I, Yee BK. The role of the endocannabinoid system in autism spectrum disorders: Evidence from mouse studies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 173:183-208. [PMID: 32711810 DOI: 10.1016/bs.pmbts.2020.04.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A substantive volume of research on autism spectrum disorder (ASD) has emerged in recent years adding to our understanding of the etiopathological process. Preclinical models in mice and rats have been highly instrumental in modeling and dissecting the contributions of a multitude of known genetic and environmental risk factors. However, the translation of preclinical data into suitable drug targets must overcome three critical hurdles: (i) ASD comprises a highly heterogeneous group of conditions that can markedly differ in terms of their clinical presentation and symptoms, (ii) the plethora of genetic and environmental risk factors suggests a complex, non-unitary, etiopathology, and (iii) the lack of consensus over the myriad of preclinical models, with respect to both construct validity and face validity. Against this backdrop, this Chapter traces how the endocannabinoid system (ECS) has emerged as a promising target for intervention with predictive validity. Recent supportive preclinical evidence is summarized, especially studies in mice demonstrating the emergence of ASD-like behaviors following diverse genetic or pharmacological manipulations targeting the ECS. The critical relevance of ECS to the complex pathogenesis of ASD is underscored by its multiple roles in modulating neuronal functions and shaping brain development. Finally, we argue that important lessons have been learned from the novel mouse models of ASD, which not only stimulate game-changing innovative treatments but also foster a consensual framework to integrate the diverse approaches applied in the search of novel treatments for ASD.
Collapse
Affiliation(s)
- Susanna Pietropaolo
- University of Bordeaux, Bordeaux Cedex, France; CNRS, INCIA, UMR 5287, Bat B2, Pessac Cedex, France.
| | - Luigi Bellocchio
- CNRS, INCIA, UMR 5287, Bat B2, Pessac Cedex, France; INSERM, U1215 NeuroCentre Magendie, Bordeaux Cedex, France
| | - Inés Bouzón-Arnáiz
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-Universitat de Barcelona), Barcelona, Spain
| | - Benjamin K Yee
- Department of Rehabilitation Sciences, Faculty of Health & Social Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
17
|
Protective effects of elevated anandamide on stress and fear-related behaviors: translational evidence from humans and mice. Mol Psychiatry 2020; 25:993-1005. [PMID: 30120421 DOI: 10.1038/s41380-018-0215-1] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 01/08/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a common, debilitating condition with limited treatment options. Extinction of fear memories through prolonged exposure therapy, the primary evidence-based behavioral treatment for PTSD, has only partial efficacy. In mice, pharmacological inhibition of fatty acid amide hydrolase (FAAH) produces elevated levels of anandamide (AEA) and promotes fear extinction, suggesting that FAAH inhibitors may aid fear extinction-based treatments. A human FAAH 385C->A substitution encodes an FAAH enzyme with reduced catabolic efficacy. Individuals homozygous for the FAAH 385A allele may therefore offer a genetic model to evaluate the impact of elevations in AEA signaling in humans, helping to inform whether FAAH inhibitors have the potential to facilitate fear extinction therapy for PTSD. To overcome the challenge posed by low frequency of the AA genotype (appr. 5%), we prospectively genotyped 423 individuals to examine the balanced groups of CC, AC, and AA individuals (n = 25/group). Consistent with its loss-of-function nature, the A allele was dose dependently associated with elevated basal AEA levels, facilitated fear extinction, and enhanced the extinction recall. Moreover, the A-allele homozygotes were protected against stress-induced decreases in AEA and negative emotional consequences of stress. In a humanized mouse model, AA homozygous mice were similarly protected against stress-induced decreases in AEA, both in the periphery, and also in the amygdala and prefrontal cortex, brain structures critically involved in fear extinction and regulation of stress responses. Collectively, these data suggest that AEA signaling can temper aspects of the stress response and that FAAH inhibition may aid the treatment for stress-related psychiatric disorders, such as PTSD.
Collapse
|
18
|
Navarrete F, García-Gutiérrez MS, Jurado-Barba R, Rubio G, Gasparyan A, Austrich-Olivares A, Manzanares J. Endocannabinoid System Components as Potential Biomarkers in Psychiatry. Front Psychiatry 2020; 11:315. [PMID: 32395111 PMCID: PMC7197485 DOI: 10.3389/fpsyt.2020.00315] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022] Open
Abstract
The high heterogeneity of psychiatric disorders leads to a lack of diagnostic precision. Therefore, the search of biomarkers is a fundamental aspect in psychiatry to reach a more personalized medicine. The endocannabinoid system (ECS) has gained increasing interest due to its involvement in many different functional processes in the brain, including the regulation of emotions, motivation, and cognition. This article reviews the role of the main components of the ECS as biomarkers in certain psychiatric disorders. Studies carried out in rodents evaluating the effects of pharmacological and genetic manipulation of cannabinoid receptors or endocannabinoids (eCBs) degrading enzymes were included. Likewise, the ECS-related alterations occurring at the molecular level in animal models reproducing some behavioral and/or neuropathological aspects of psychiatric disorders were reviewed. Furthermore, clinical studies evaluating gene or protein alterations in post-mortem brain tissue or in vivo blood, plasma, and cerebrospinal fluid (CSF) samples were analyzed. Also, the results from neuroimaging studies using positron emission tomography (PET) or functional magnetic resonance (fMRI) were included. This review shows the close involvement of cannabinoid receptor 1 (CB1r) in stress regulation and the development of mood disorders [anxiety, depression, bipolar disorder (BD)], in post-traumatic stress disorder (PTSD), as well as in the etiopathogenesis of schizophrenia, attention deficit hyperactivity disorder (ADHD), or eating disorders (i.e. anorexia and bulimia nervosa). On the other hand, recent results reveal the potential therapeutic action of the endocannabinoid tone manipulation by inhibition of eCBs degrading enzymes, as well as by the modulation of cannabinoid receptor 2 (CB2r) activity on anxiolytic, antidepressive, or antipsychotic associated effects. Further clinical research studies are needed; however, current evidence suggests that the components of the ECS may become promising biomarkers in psychiatry to improve, at least in part, the diagnosis and pharmacological treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Rosa Jurado-Barba
- Instituto de Investigación i+12, Hospital Universitario 12 de Octubre, Madrid, Spain.,Servicio de Psiquiatría, Hospital Universitario 12 de Octubre, Madrid, Spain.,Departamento de Psicología, Facultad de Educación y Salud, Universidad Camilo José Cela, Madrid, Spain
| | - Gabriel Rubio
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.,Instituto de Investigación i+12, Hospital Universitario 12 de Octubre, Madrid, Spain.,Servicio de Psiquiatría, Hospital Universitario 12 de Octubre, Madrid, Spain.,Department of Psychiatry, Complutense University of Madrid, Madrid, Spain
| | - Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | | | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| |
Collapse
|
19
|
Endocannabinoids, stress signaling, and the locus coeruleus-norepinephrine system. Neurobiol Stress 2019; 11:100176. [PMID: 31236436 PMCID: PMC6582240 DOI: 10.1016/j.ynstr.2019.100176] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/23/2019] [Accepted: 05/23/2019] [Indexed: 01/14/2023] Open
Abstract
The endocannabinoid (eCB) system has been implicated in a variety of physiological functions due to abundant expression of its receptors and endogenous ligands in the central nervous system. Substantial progress has been made in understanding how the eCB system influences the brain norepinephrine (NE) system, an important neurochemical target in the continued development of new therapies for stress-induced psychiatric disorders. We, and others, have characterized the neuroanatomical, biochemical and pharmacological effects of cannabinoid receptor modulation on brain noradrenergic circuitry and defined how molecular elements of the eCB system are positioned to directly impact the locus coeruleus (LC)-prefrontal cortex pathway, a neural circuit well recognized for contributing to symptoms of hyperarousal, a key pathophysiological feature of stress-related disorders. We also described molecular and electrophysiological properties of LC noradrenergic neurons and NE release in the medial prefrontal cortex under conditions of cannabinoid type 1 receptor deletion. Finally, we identified how stress influences cannabinoid modulation of the coeruleo-cortical pathway. A number of significant findings emerged from these studies that will be summarized in the present review and have important implications for clinical studies targeting the eCB system in the treatment of stress-induced psychiatric disorders.
Collapse
|
20
|
Endocannabinoid system, stress and HPA axis. Eur J Pharmacol 2018; 834:230-239. [DOI: 10.1016/j.ejphar.2018.07.039] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/19/2018] [Indexed: 11/19/2022]
|
21
|
Gorzkiewicz A, Szemraj J. Brain endocannabinoid signaling exhibits remarkable complexity. Brain Res Bull 2018; 142:33-46. [PMID: 29953913 DOI: 10.1016/j.brainresbull.2018.06.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/06/2018] [Accepted: 06/21/2018] [Indexed: 01/04/2023]
Abstract
The endocannabinoid (eCB) signaling system is one of the most extensive of the mammalian brain. Despite the involvement of only few specific ligands and receptors, the system encompasses a vast diversity of triggered mechanisms and driven effects. It mediates a wide range of phenomena, including the regulation of transmitter release, neural excitability, synaptic plasticity, impulse spread, long-term neuronal potentiation, neurogenesis, cell death, lineage segregation, cell migration, inflammation, oxidative stress, nociception and the sleep cycle. It is also known to be involved in the processes of learning and memory formation. This extensive scope of action is attained by combining numerous variables. In a properly functioning brain, the correlations of these variables are kept in a strictly controlled balance; however, this balance is disrupted in many pathological conditions. However, while this balance is known to be disrupted by drugs in the case of addicts, the stimuli and mechanisms influencing the neurodegenerating brain remain elusive. This review examines the multiple factors and phenomena affecting the eCB signaling system in the brain. It evaluates techniques of controlling the eCB system to identify the obstacles in their applications and highlights the crucial interdependent variables that may influence biomedical research outcomes.
Collapse
Affiliation(s)
- Anna Gorzkiewicz
- Medical University of Lodz, ul.Mazowiecka 6/8, 92-215, Lodz, Poland.
| | - Janusz Szemraj
- Medical University of Lodz, ul.Mazowiecka 6/8, 92-215, Lodz, Poland
| |
Collapse
|
22
|
Circulating endocannabinoids and affect regulation in human subjects. Psychoneuroendocrinology 2018; 92:66-71. [PMID: 29627714 DOI: 10.1016/j.psyneuen.2018.03.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 12/31/2022]
Abstract
The endocannabinoid (EC) system influences a wide variety of neurobiological processes including affect and emotionality as well as other neuropsychiatric functions. In this study we examined the relationship of circulating endocannabinoids [anandamide (AEA) and 2-arachidonoylglycerol (2-AG)] with affect and emotionality in 175 individuals with (n = 115) and without (n = 60) mood, anxiety, and/or personality disorders. Circulating AEA levels displayed a modest, though statistically significant, inverse relationship with a composite measure of affect regulation (β = - 0.264, p = 0.009), due to its relationship with affect intensity (β = - 0.225, p = 0.021) across all study participants. Neither AEA nor 2-AG level differed as a function of any syndromal/personality disorder and neither correlated significantly with state depression or state anxiety scores. These data suggest that circulating levels endocannabinoids may play a role in emotionality across individuals regardless of defined psychiatric disorder.
Collapse
|
23
|
Fogaça MV, Campos AC, Coelho LD, Duman RS, Guimarães FS. 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. [PMID: 29510186 DOI: 10.1016/j.neuropharm.2018.03.001] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/29/2018] [Accepted: 03/01/2018] [Indexed: 11/30/2022]
Abstract
Repeated injections of cannabidiol (CBD), the major non-psychotomimetic compound present in the Cannabis sativa plant, attenuate the anxiogenic effects induced by Chronic Unpredictable Stress (CUS). The specific mechanisms remain to be fully understood but seem to involve adult hippocampal neurogenesis and recruitment of endocannabinoids. Here we investigated for the first time if the behavioral and pro-neurogenic effects of CBD administered concomitant the CUS procedure (14 days) are mediated by CB1, CB2 or 5HT1A receptors, as well as CBD effects on dendritic remodeling and on intracellular/synaptic signaling (fatty acid amide hydrolase - FAAH, Akt, GSK3β and the synaptic proteins Synapsin Ia/b, mGluR1 and PSD95). After 14 days, CBD injections (30 mg/kg) induced anxiolytic responses in stressed animals in the elevated plus-maze and novelty suppressed feeding tests, that were blocked by pre-treatment with a CB1 (AM251, 0.3 mg/kg) or CB2 (AM630, 0.3 mg/kg), but not by a 5HT1A (WAY100635, 0.05 mg/kg) receptor antagonist. Golgi staining and immunofluorescence revealed that these effects were associated with an increase in hippocampal neurogenesis and spine density in the dentate gyrus of the hippocampus. AM251 and AM630 abolished the effects of CBD on spines density. However, AM630 was more effective in attenuating the pro-neurogenic effects of CBD. CBD decreased FAAH and increased p-GSK3β expression in stressed animals, which was also attenuated by AM630. These results indicate that CBD prevents the behavioral effects caused by CUS probably due to a facilitation of endocannabinoid neurotransmission and consequent CB1/CB2 receptors activation, which could recruit intracellular/synaptic proteins involved in neurogenesis and dendritic remodeling.
Collapse
Affiliation(s)
- Manoela V Fogaça
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP-USP), Bandeirantes Avenue 3900, 14049-900, Ribeirão Preto, São Paulo, Brazil; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo (USP), Brazil; Department of Psychiatry, Yale University School of Medicine, 34 Park Street 06520, New Haven, CT, United States.
| | - Alline C Campos
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP-USP), Bandeirantes Avenue 3900, 14049-900, Ribeirão Preto, São Paulo, Brazil; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo (USP), Brazil
| | - Ludmila D Coelho
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP-USP), Bandeirantes Avenue 3900, 14049-900, Ribeirão Preto, São Paulo, Brazil; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo (USP), Brazil
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street 06520, New Haven, CT, United States
| | - Francisco S Guimarães
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP-USP), Bandeirantes Avenue 3900, 14049-900, Ribeirão Preto, São Paulo, Brazil; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo (USP), Brazil
| |
Collapse
|
24
|
Integrating Endocannabinoid Signaling and Cannabinoids into the Biology and Treatment of Posttraumatic Stress Disorder. Neuropsychopharmacology 2018; 43:80-102. [PMID: 28745306 PMCID: PMC5719095 DOI: 10.1038/npp.2017.162] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/17/2017] [Accepted: 07/20/2017] [Indexed: 01/21/2023]
Abstract
Exposure to stress is an undeniable, but in most cases surmountable, part of life. However, in certain individuals, exposure to severe or cumulative stressors can lead to an array of pathological conditions including posttraumatic stress disorder (PTSD), characterized by debilitating trauma-related intrusive thoughts, avoidance behaviors, hyperarousal, as well as depressed mood and anxiety. In the context of the rapidly changing political and legal landscape surrounding use of cannabis products in the USA, there has been a surge of public and research interest in the role of cannabinoids in the regulation of stress-related biological processes and in their potential therapeutic application for stress-related psychopathology. Here we review the current state of knowledge regarding the effects of cannabis and cannabinoids in PTSD and the preclinical and clinical literature on the effects of cannabinoids and endogenous cannabinoid signaling systems in the regulation of biological processes related to the pathogenesis of PTSD. Potential therapeutic implications of the reviewed literature are also discussed. Finally, we propose that a state of endocannabinoid deficiency could represent a stress susceptibility endophenotype predisposing to the development of trauma-related psychopathology and provide biologically plausible support for the self-medication hypotheses used to explain high rates of cannabis use in patients with trauma-related disorders.
Collapse
|
25
|
Leonard MZ, Alapafuja SO, Ji L, Shukla VG, Liu Y, Nikas SP, Makriyannis A, Bergman J, Kangas BD. Cannabinoid CB 1 Discrimination: Effects of Endocannabinoids and Catabolic Enzyme Inhibitors. J Pharmacol Exp Ther 2017; 363:314-323. [PMID: 28947487 PMCID: PMC5683067 DOI: 10.1124/jpet.117.244392] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/14/2017] [Indexed: 11/22/2022] Open
Abstract
An improved understanding of the endocannabinoid system has provided new avenues of drug discovery and development toward the management of pain and other behavioral maladies. Exogenous cannabinoid type 1 (CB1) receptor agonists such as Δ9-tetrahydrocannabinol are increasingly used for their medicinal actions; however, their utility is constrained by concern regarding abuse-related subjective effects. This has led to growing interest in the clinical benefit of indirectly enhancing the activity of the highly labile endocannabinoids N-arachidonoylethanolamine [AEA (or anandamide)] and/or 2-arachidonoylglycerol (2-AG) via catabolic enzyme inhibition. The present studies were conducted to determine whether such actions can lead to CB1 agonist-like subjective effects, as reflected in CB1-related discriminative stimulus effects in laboratory subjects. Squirrel monkeys (n = 8) that discriminated the CB1 full agonist AM4054 (0.01 mg/kg) from vehicle were used to study, first, the inhibitors of fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MGL) alone or in combination [FAAH (URB597, AM4303); MGL (AM4301); FAAH/MGL (JZL195, AM4302)] and, second, the ability of the endocannabinoids AEA and 2-AG to produce CB1 agonist-like effects when administered alone or after enzyme inhibition. Results indicate that CB1-related discriminative stimulus effects were produced by combined, but not selective, inhibition of FAAH and MGL, and that these effects were nonsurmountably antagonized by low doses of rimonabant. Additionally, FAAH or MGL inhibition revealed CB1-like subjective effects produced by AEA but not by 2-AG. Taken together, the present data suggest that therapeutic effects of combined, but not selective, enhancement of AEA or 2-AG activity via enzyme inhibition may be accompanied by CB1 receptor-mediated subjective effects.
Collapse
MESH Headings
- Adamantane/administration & dosage
- Adamantane/adverse effects
- Adamantane/analogs & derivatives
- Adamantane/pharmacology
- Amidohydrolases/antagonists & inhibitors
- Amidohydrolases/metabolism
- Animals
- Arachidonic Acids/administration & dosage
- Arachidonic Acids/agonists
- Arachidonic Acids/antagonists & inhibitors
- Arachidonic Acids/pharmacology
- Behavior, Animal/drug effects
- Cannabinoid Receptor Antagonists/administration & dosage
- Cannabinoid Receptor Antagonists/adverse effects
- Cannabinoid Receptor Antagonists/pharmacology
- Cannabinol/administration & dosage
- Cannabinol/adverse effects
- Cannabinol/analogs & derivatives
- Cannabinol/pharmacology
- Discrimination Learning/drug effects
- Dose-Response Relationship, Drug
- Drug Agonism
- Drug Antagonism
- Drugs, Investigational/administration & dosage
- Drugs, Investigational/adverse effects
- Drugs, Investigational/pharmacology
- Endocannabinoids/administration & dosage
- Endocannabinoids/agonists
- Endocannabinoids/antagonists & inhibitors
- Endocannabinoids/pharmacology
- Enzyme Inhibitors/administration & dosage
- Enzyme Inhibitors/adverse effects
- Enzyme Inhibitors/pharmacology
- Glycerides/administration & dosage
- Glycerides/agonists
- Glycerides/antagonists & inhibitors
- Glycerides/pharmacology
- Injections, Intramuscular
- Injections, Intravenous
- Ligands
- Male
- Monoacylglycerol Lipases/antagonists & inhibitors
- Monoacylglycerol Lipases/metabolism
- Nerve Tissue Proteins/agonists
- Nerve Tissue Proteins/antagonists & inhibitors
- Nerve Tissue Proteins/metabolism
- Polyunsaturated Alkamides
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Saimiri
Collapse
Affiliation(s)
- Michael Z Leonard
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Shakiru O Alapafuja
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Lipin Ji
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Vidyanand G Shukla
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Yingpeng Liu
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Spyros P Nikas
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Alexandros Makriyannis
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Jack Bergman
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Brian D Kangas
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| |
Collapse
|
26
|
Natividad LA, Buczynski MW, Herman MA, Kirson D, Oleata CS, Irimia C, Polis I, Ciccocioppo R, Roberto M, Parsons LH. Constitutive Increases in Amygdalar Corticotropin-Releasing Factor and Fatty Acid Amide Hydrolase Drive an Anxious Phenotype. Biol Psychiatry 2017; 82:500-510. [PMID: 28209423 PMCID: PMC5509512 DOI: 10.1016/j.biopsych.2017.01.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/21/2016] [Accepted: 01/04/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND Corticotropin-releasing factor (CRF) mediates anxiogenic responses by activating CRF type 1 (CRF1) receptors in limbic brain regions. Anxiety is further modulated by the endogenous cannabinoid (eCB) system that attenuates the synaptic effects of stress. In the amygdala, acute stress activates the enzymatic clearance of the eCB N-arachidonoylethanolamine via fatty acid amide hydrolase (FAAH), although it is unclear whether chronic dysregulation of CRF systems induces maladaptive changes in amygdalar eCB signaling. Here, we used genetically selected Marchigian Sardinian P (msP) rats carrying an innate overexpression of CRF1 receptors to study the role of constitutive upregulation in CRF systems on amygdalar eCB function and persistent anxiety-like effects. METHODS We applied behavioral, pharmacological, and biochemical methods to broadly characterize anxiety-like behaviors and amygdalar eCB clearance enzymes in msP versus nonselected Wistar rats. Subsequent studies examined the influence of dysregulated CRF and FAAH systems in altering excitatory transmission in the central amygdala (CeA). RESULTS msPs display an anxious phenotype accompanied by elevations in amygdalar FAAH activity and reduced dialysate N-arachidonoylethanolamine levels in the CeA. Elevations in CRF-CRF1 signaling dysregulate FAAH activity, and this genotypic difference is normalized with pharmacological blockade of CRF1 receptors. msPs also exhibit elevated baseline glutamatergic transmission in the CeA, and dysregulated CRF-FAAH facilitates stress-induced increases in glutamatergic activity. Treatment with an FAAH inhibitor relieves sensitized glutamatergic responses in msPs and attenuates the anxiety-like phenotype. CONCLUSIONS Pathological anxiety and stress hypersensitivity are driven by constitutive increases in CRF1 signaling that dysregulate N-arachidonoylethanolamine signaling mechanisms and reduce neuronal inhibitory control of CeA glutamatergic synapses.
Collapse
Affiliation(s)
- Luis A Natividad
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California
| | - Matthew W Buczynski
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California; Virginia Tech School of Neuroscience, Virginia Tech, Blacksburg, Virginia
| | - Melissa A Herman
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California
| | - Dean Kirson
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California
| | - Christopher S Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California
| | - Cristina Irimia
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California
| | - Ilham Polis
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California.
| | - Loren H Parsons
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
27
|
Mandolesi G, Bullitta S, Fresegna D, Gentile A, De Vito F, Dolcetti E, Rizzo FR, Strimpakos G, Centonze D, Musella A. Interferon-γ causes mood abnormalities by altering cannabinoid CB1 receptor function in the mouse striatum. Neurobiol Dis 2017; 108:45-53. [PMID: 28757328 DOI: 10.1016/j.nbd.2017.07.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/21/2017] [Accepted: 07/26/2017] [Indexed: 01/10/2023] Open
Abstract
Interferon-γ (IFN-γ) has been implicated in the pathogenesis of multiple sclerosis (MS) and in its animal model, experimental autoimmune encephalomyelitis (EAE). The type-1 cannabinoid receptors (CB1Rs) are heavily involved in MS pathophysiology, and a growing body of evidence suggests that mood disturbances reflect specific effects of proinflammatory cytokines on neuronal activity. Here, we investigated whether IFN-γ could exert a role in the anxiety- and depressive-like behavior observed in mice with EAE, and in the modulation of CB1Rs. Anxiety and depression in fact are often diagnosed in MS, and have already been shown to depend on cannabinoid system. We performed biochemical, behavioral and electrophysiological experiments to assess the role of IFN-γ on mood control and on synaptic transmission in mice. Intracerebroventricular delivery of IFN-γ caused a depressive- and anxiety-like behavior in mice, associated with the selective dysfunction of CB1Rs controlling GABA transmission in the striatum. EAE induction was associated with increased striatal expression of IFN-γ, and with CB1R transmission deficits, which were rescued by pharmacological blockade of IFN-γ. IFN-γ was unable to replicate the effects of EAE on excitatory and inhibitory transmission in the striatum, but mimicked the effects of EAE on CB1R function in this brain area. Overall these results indicate that IFN-γ exerts a relevant control on mood, through the modulation of CB1R function. A better understanding of the biological pathways underling the psychological disorders during neuroinflammatory conditions is crucial for developing effective therapeutic strategies.
Collapse
Affiliation(s)
- Georgia Mandolesi
- Centro Europeo per la Ricerca sul Cervello (CERC), IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Silvia Bullitta
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - Diego Fresegna
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - Antonietta Gentile
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - Francesca De Vito
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - Ettore Dolcetti
- Centro Europeo per la Ricerca sul Cervello (CERC), IRCCS Fondazione Santa Lucia, 00143 Rome, Italy; Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
| | - Francesca R Rizzo
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - Georgios Strimpakos
- Institute of Cell Biology and Neurobiology CNR, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy.
| | - Alessandra Musella
- Centro Europeo per la Ricerca sul Cervello (CERC), IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| |
Collapse
|
28
|
Shoshan N, Segev A, Abush H, Mizrachi Zer-Aviv T, Akirav I. Cannabinoids prevent the differential long-term effects of exposure to severe stress on hippocampal- and amygdala-dependent memory and plasticity. Hippocampus 2017; 27:1093-1109. [DOI: 10.1002/hipo.22755] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/22/2017] [Accepted: 06/20/2017] [Indexed: 11/05/2022]
Affiliation(s)
- Noa Shoshan
- Department of Psychology; University of Haifa; Haifa 3498838 Israel
| | - Amir Segev
- Department of Psychology; University of Haifa; Haifa 3498838 Israel
| | - Hila Abush
- Department of Psychology; University of Haifa; Haifa 3498838 Israel
| | | | - Irit Akirav
- Department of Psychology; University of Haifa; Haifa 3498838 Israel
| |
Collapse
|
29
|
Henricks AM, Berger AL, Lugo JM, Baxter-Potter LN, Bieniasz KV, Petrie G, Sticht MA, Hill MN, McLaughlin RJ. Sex- and hormone-dependent alterations in alcohol withdrawal-induced anxiety and corticolimbic endocannabinoid signaling. Neuropharmacology 2017; 124:121-133. [PMID: 28554848 DOI: 10.1016/j.neuropharm.2017.05.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/04/2017] [Accepted: 05/23/2017] [Indexed: 01/01/2023]
Abstract
Alcohol dependence is associated with anxiety during withdrawal. The endocannabinoid (ECB) system participates in the neuroendocrine and behavioral response to stress and changes in corticolimbic ECB signaling may contribute to alcohol withdrawal-induced anxiety. Moreover, symptoms of alcohol withdrawal differ between sexes and sexual dimorphism in withdrawal-induced ECB recruitment may be a contributing factor. Herein, we exposed intact male and female rats and ovariectomized (OVX) female rats with or without estradiol (E2) replacement to 6 weeks of chronic intermittent alcohol vapor and measured anxiety-like behavior, ECB content, and ECB-related mRNA in the basolateral amygdala (BLA) and ventromedial prefrontal cortex (vmPFC). Acute alcohol withdrawal increased anxiety-like behavior, produced widespread disturbances in ECB-related mRNA, and reduced anandamide (AEA) content in the BLA and 2-arachidonoylglycerol (2-AG) content in the vmPFC of male, but not female rats. Similar to males, alcohol-exposed OVX females showed reductions in Napepld mRNA in the BLA, decreased AEA content in the BLA and vmPFC, and reductions in all ECB-related genes measured in the vmPFC. Importantly, E2 replacement prevented withdrawal-induced alterations in ECB content (but not mRNA) in OVX females, and although alcohol-exposed OVX females failed to exhibit more anxiety compared to their respective control, chronic alcohol exposure abolished the anxiolytic properties of E2 in OVX rats. These data indicate that ovarian sex hormones (but not E2 alone) protect against withdrawal-induced alterations in corticolimbic ECB signaling but do not impart resilience to withdrawal-induced anxiety. Thus, the mechanisms implicated in the manifestation of alcohol withdrawal-induced anxiety are most likely sex-specific. This article is part of the Special Issue entitled "A New Dawn in Cannabinoid Neurobiology".
Collapse
Affiliation(s)
- Angela M Henricks
- Department of Psychology, Washington State University, Pullman, WA 99164, USA
| | - Anthony L Berger
- Department of Psychology, Washington State University, Pullman, WA 99164, USA
| | - Janelle M Lugo
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Lydia N Baxter-Potter
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Kennedy V Bieniasz
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Gavin Petrie
- Hotchkiss Brain Institute, Departments of Cell Biology and Anatomy and Psychiatry, University of Calgary, Calgary, Alberta, Canada
| | - Martin A Sticht
- Hotchkiss Brain Institute, Departments of Cell Biology and Anatomy and Psychiatry, University of Calgary, Calgary, Alberta, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, Departments of Cell Biology and Anatomy and Psychiatry, University of Calgary, Calgary, Alberta, Canada
| | - Ryan J McLaughlin
- Department of Psychology, Washington State University, Pullman, WA 99164, USA; Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA 99164, USA; Translational Addiction Research Center, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
30
|
Patel S, Hill MN, Cheer JF, Wotjak CT, Holmes A. The endocannabinoid system as a target for novel anxiolytic drugs. Neurosci Biobehav Rev 2017; 76:56-66. [PMID: 28434588 PMCID: PMC5407316 DOI: 10.1016/j.neubiorev.2016.12.033] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/22/2016] [Accepted: 12/16/2016] [Indexed: 12/01/2022]
Abstract
The endocannabinoid (eCB) system has attracted attention for its role in various behavioral and brain functions, and as a therapeutic target in neuropsychiatric disease states, including anxiety disorders and other conditions resulting from dysfunctional responses to stress. In this mini-review, we highlight components of the eCB system that offer potential 'druggable' targets for new anxiolytic medications, emphasizing some of the less well-discussed options. We discuss how selectively amplifying eCBs recruitment by interfering with eCB-degradation, via fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), has been linked to reductions in anxiety-like behaviors in rodents and variation in human anxiety symptoms. We also discuss a non-canonical route to regulate eCB degradation that involves interfering with cyclooxygenase-2 (COX-2). Next, we discuss approaches to targeting eCB receptor-signaling in ways that do not involve the cannabinoid receptor subtype 1 (CB1R); by targeting the CB2R subtype and the transient receptor potential vanilloid type 1 (TRPV1). Finally, we review evidence that cannabidiol (CBD), while representing a less specific pharmacological approach, may be another way to modulate eCBs and interacting neurotransmitter systems to alleviate anxiety. Taken together, these various approaches provide a range of plausible paths to developing novel compounds that could prove useful for treating trauma-related and anxiety disorders.
Collapse
Affiliation(s)
- Sachin Patel
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, USA; Vanderbilt Kennedy Center for Human Development, Vanderbilt University Medical Center, Nashville, USA
| | - Mathew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada; Departments of Cell Biology and Anatomy and Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology and Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Carsten T Wotjak
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology & Neurogenetics, Munich, Germany
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
31
|
Chronic stress leads to epigenetic dysregulation in the neuropeptide-Y and cannabinoid CB1 receptor genes in the mouse cingulate cortex. Neuropharmacology 2017; 113:301-313. [DOI: 10.1016/j.neuropharm.2016.10.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 10/06/2016] [Accepted: 10/08/2016] [Indexed: 12/16/2022]
|
32
|
Morena M, Leitl KD, Vecchiarelli HA, Gray JM, Campolongo P, Hill MN. Emotional arousal state influences the ability of amygdalar endocannabinoid signaling to modulate anxiety. Neuropharmacology 2016; 111:59-69. [DOI: 10.1016/j.neuropharm.2016.08.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/26/2016] [Accepted: 08/16/2016] [Indexed: 01/04/2023]
|
33
|
Cannabinoids and post-traumatic stress disorder: clinical and preclinical evidence for treatment and prevention. Behav Pharmacol 2016; 27:561-9. [DOI: 10.1097/fbp.0000000000000253] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
34
|
Spagnolo PA, Ramchandani VA, Schwandt ML, Kwako LE, George DT, Mayo LM, Hillard CJ, Heilig M. FAAH Gene Variation Moderates Stress Response and Symptom Severity in Patients with Posttraumatic Stress Disorder and Comorbid Alcohol Dependence. Alcohol Clin Exp Res 2016; 40:2426-2434. [PMID: 27716956 DOI: 10.1111/acer.13210] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/30/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND A common single nucleotide polymorphism (C385A) in the human fatty acid amide hydrolase (FAAH) gene has been associated with decreased distress responses in healthy volunteers, but its role in psychiatric disorders remains unknown. Here, we obtained genotypes and carried out a secondary analysis of subjects from a trial of comorbid posttraumatic stress disorder (PTSD) and alcohol dependence (AD). We evaluated the effects of C385A variation on behavioral and biochemical biomarkers of distress responses. METHODS Forty-nine patients with PTSD and AD were admitted for 4 weeks to an experimental medicine unit at the National Institutes of Health Clinical Center. Following detoxification, stress reactivity and peripheral endocannabinoid (eCB) levels were assessed in response to a challenge session using personalized auditory guided imagery. Over the course of the study, subjects were also evaluated for changes in PTSD symptom severity. RESULTS FAAH C385A allele carriers showed a marked increase in serum anandamide levels at baseline and throughout the stress challenge procedure compared with C allele homozygotes, while levels of eCBs primarily metabolized through other enzymatic activity, such as 2-arachidonoylglycerol, did not differ between genotype groups. FAAH C385A carriers also had decreased subjective anxiety responses to the stress challenge. Similar effects of FAAH C385A genotype were found at the level of clinical PTSD symptom severity, in particular in the arousal domain. CONCLUSIONS This is to our knowledge the first study showing that FAAH C385A variation modulates stress responses in subjects with disorders characterized by increased stress reactivity. These findings point to the eCB pathway as a promising target for future antistress therapeutics.
Collapse
Affiliation(s)
- Primavera A Spagnolo
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Vijay A Ramchandani
- Section on Human Psychopharmacology, Division of Intramural Clinical and Biomedical Research, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Melanie L Schwandt
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Laura E Kwako
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - David T George
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Leah M Mayo
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Markus Heilig
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| |
Collapse
|
35
|
Tomas-Roig J, Piscitelli F, Gil V, del Río J, Moore T, Agbemenyah H, Salinas-Riester G, Pommerenke C, Lorenzen S, Beißbarth T, Hoyer-Fender S, Di Marzo V, Havemann-Reinecke U. Social defeat leads to changes in the endocannabinoid system: An overexpression of calreticulin and motor impairment in mice. Behav Brain Res 2016; 303:34-43. [DOI: 10.1016/j.bbr.2016.01.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/13/2016] [Accepted: 01/16/2016] [Indexed: 12/12/2022]
|
36
|
Sustained glucocorticoid exposure recruits cortico-limbic CRH signaling to modulate endocannabinoid function. Psychoneuroendocrinology 2016; 66:151-8. [PMID: 26821211 PMCID: PMC4788523 DOI: 10.1016/j.psyneuen.2016.01.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/09/2015] [Accepted: 01/05/2016] [Indexed: 12/21/2022]
Abstract
Sustained exposure to stress or corticosteroids is known to cause changes in brain endocannabinoid (eCB) signaling, such that tissue contents of the eCBs N-arachidonylethanolamine (AEA) are generally reduced while 2-arachidonoylglycerol (2-AG) levels increase. These changes in eCB signaling are important for many of the aspects of chronic stress, such as anxiety, reward sensitivity and stress adaptation, yet the mechanisms mediating these changes are not fully understood. We have recently found that the stress-related neuropeptide corticotropin-releasing hormone (CRH), acting through the CRH type 1 receptor (CRHR1), can reduce AEA content by increasing its hydrolysis by the enzyme fatty acid amide hydrolase (FAAH) as well as increase 2-AG contents. As extra-hypothalamic CRH is upregulated by chronic corticosteroid or stress exposure, we hypothesized that increased CRH signaling through CRHR1 contributes to the effects of chronic corticosteroid exposure on the eCB system within the amygdala and prefrontal cortex. Male rats were exposed to 7 days of systemic corticosterone capsules, with or without concurrent exposure to a CRHR1 antagonist, after which we examined eCB content. Consistent with previous studies in the amygdala, sustained corticosterone exposure increases CRH mRNA in the prefrontal cortex. As was shown previously, FAAH activity was increased and AEA contents were reduced within the amygdala and prefrontal cortex following chronic corticosterone exposure. Chronic corticosterone exposure also elevated 2-AG content in the prefrontal cortex but not the amygdala. These corticosteroid-driven changes were all blocked by systemic CRHR1 antagonism. Consistent with these data indicating sustained increases in CRH signaling can mediate the effects of chronic elevations in corticosteroids, CRH overexpressing mice also exhibited increased FAAH-mediated AEA hydrolysis in the amygdala and prefrontal cortex compared to wild type. CRH overexpression increased 2-AG content in the amygdala, but not the prefrontal cortex. These data indicate that chronic elevations in CRH signaling, as is seen following exposure to chronic elevations in corticosterone or stress, drive persistent changes in eCB function. As reductions in AEA signaling mediate the effects of CRH and chronic stress on anxiety, these data provide a mechanism linking these processes.
Collapse
|
37
|
Morena M, Patel S, Bains JS, Hill MN. Neurobiological Interactions Between Stress and the Endocannabinoid System. Neuropsychopharmacology 2016; 41:80-102. [PMID: 26068727 PMCID: PMC4677118 DOI: 10.1038/npp.2015.166] [Citation(s) in RCA: 432] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 05/20/2015] [Accepted: 05/20/2015] [Indexed: 12/18/2022]
Abstract
Stress affects a constellation of physiological systems in the body and evokes a rapid shift in many neurobehavioral processes. A growing body of work indicates that the endocannabinoid (eCB) system is an integral regulator of the stress response. In the current review, we discuss the evidence to date that demonstrates stress-induced regulation of eCB signaling and the consequential role changes in eCB signaling have with respect to many of the effects of stress. Across a wide array of stress paradigms, studies have generally shown that stress evokes bidirectional changes in the two eCB molecules, anandamide (AEA) and 2-arachidonoyl glycerol (2-AG), with stress exposure reducing AEA levels and increasing 2-AG levels. Additionally, in almost every brain region examined, exposure to chronic stress reliably causes a downregulation or loss of cannabinoid type 1 (CB1) receptors. With respect to the functional role of changes in eCB signaling during stress, studies have demonstrated that the decline in AEA appears to contribute to the manifestation of the stress response, including activation of the hypothalamic-pituitary-adrenal (HPA) axis and increases in anxiety behavior, while the increased 2-AG signaling contributes to termination and adaptation of the HPA axis, as well as potentially contributing to changes in pain perception, memory and synaptic plasticity. More so, translational studies have shown that eCB signaling in humans regulates many of the same domains and appears to be a critical component of stress regulation, and impairments in this system may be involved in the vulnerability to stress-related psychiatric conditions, such as depression and posttraumatic stress disorder. Collectively, these data create a compelling argument that eCB signaling is an important regulatory system in the brain that largely functions to buffer against many of the effects of stress and that dynamic changes in this system contribute to different aspects of the stress response.
Collapse
Affiliation(s)
- Maria Morena
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada
| | - Sachin Patel
- Department of Molecular Physiology and Biophysics and Psychiatry, Vanderbilt Brain Institute, Vanderbilt-Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jaideep S Bains
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada,Departments of Cell Biology and Anatomy and Psychiatry, University of Calgary, Calgary, AB, Canada,Departments of Cell Biology and Anatomy, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N4N1, Canada, Tel: +1 403 220 8466, Fax: +1 403 283 2700, E-mail:
| |
Collapse
|
38
|
McEwen BS, Bowles NP, Gray JD, Hill MN, Hunter RG, Karatsoreos IN, Nasca C. Mechanisms of stress in the brain. Nat Neurosci 2015; 18:1353-63. [PMID: 26404710 PMCID: PMC4933289 DOI: 10.1038/nn.4086] [Citation(s) in RCA: 920] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 07/08/2015] [Indexed: 02/07/2023]
Abstract
The brain is the central organ involved in perceiving and adapting to social and physical stressors via multiple interacting mediators, from the cell surface to the cytoskeleton to epigenetic regulation and nongenomic mechanisms. A key result of stress is structural remodeling of neural architecture, which may be a sign of successful adaptation, whereas persistence of these changes when stress ends indicates failed resilience. Excitatory amino acids and glucocorticoids have key roles in these processes, along with a growing list of extra- and intracellular mediators that includes endocannabinoids and brain-derived neurotrophic factor (BDNF). The result is a continually changing pattern of gene expression mediated by epigenetic mechanisms involving histone modifications and CpG methylation and hydroxymethylation as well as by the activity of retrotransposons that may alter genomic stability. Elucidation of the underlying mechanisms of plasticity and vulnerability of the brain provides a basis for understanding the efficacy of interventions for anxiety and depressive disorders as well as age-related cognitive decline.
Collapse
Affiliation(s)
- Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York, USA
| | - Nicole P Bowles
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York, USA
| | - Jason D Gray
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York, USA
| | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Richard G Hunter
- Department of Psychology, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - Ilia N Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Carla Nasca
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
39
|
Fitzgibbon M, Finn DP, Roche M. High Times for Painful Blues: The Endocannabinoid System in Pain-Depression Comorbidity. Int J Neuropsychopharmacol 2015; 19:pyv095. [PMID: 26342110 PMCID: PMC4815466 DOI: 10.1093/ijnp/pyv095] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/17/2015] [Indexed: 01/06/2023] Open
Abstract
Depression and pain are two of the most debilitating disorders worldwide and have an estimated cooccurrence of up to 80%. Comorbidity of these disorders is more difficult to treat, associated with significant disability and impaired health-related quality of life than either condition alone, resulting in enormous social and economic cost. Several neural substrates have been identified as potential mediators in the association between depression and pain, including neuroanatomical reorganization, monoamine and neurotrophin depletion, dysregulation of the hypothalamo-pituitary-adrenal axis, and neuroinflammation. However, the past decade has seen mounting evidence supporting a role for the endogenous cannabinoid (endocannabinoid) system in affective and nociceptive processing, and thus, alterations in this system may play a key role in reciprocal interactions between depression and pain. This review will provide an overview of the preclinical evidence supporting an interaction between depression and pain and the evidence supporting a role for the endocannabinoid system in this interaction.
Collapse
Affiliation(s)
| | | | - Michelle Roche
- Physiology (Ms Fitzgibbon and Dr Roche), and Pharmacology and Therapeutics (Dr Finn), School of Medicine, Galway Neuroscience Centre and Centre for Pain Research (Ms Fitzgibbon, Dr Finn, and Dr Roche), National Centre for Biomedical Engineering Science, National University of Ireland Galway, Ireland.
| |
Collapse
|
40
|
Vázquez C, Tolón RM, Grande MT, Caraza M, Moreno M, Koester EC, Villaescusa B, Ruiz-Valdepeñas L, Fernández-Sánchez FJ, Cravatt BF, Hillard CJ, Romero J. Endocannabinoid regulation of amyloid-induced neuroinflammation. Neurobiol Aging 2015; 36:3008-3019. [PMID: 26362942 DOI: 10.1016/j.neurobiolaging.2015.08.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 07/20/2015] [Accepted: 08/04/2015] [Indexed: 12/26/2022]
Abstract
The modulation of endocannabinoid (EC) levels and the activation of cannabinoid receptors are seen as promising therapeutic strategies in a variety of diseases, including Alzheimer's disease (AD). We aimed to evaluate the effect of the pharmacologic and genetic inhibition of anandamide-degrading enzyme in a mouse model of AD (5xFAD). Pharmacologic inhibition of the fatty acid amide hydrolase (FAAH) had little impact on the expression of key enzymes and cytokines and also on the cognitive impairment, plaque deposition, and gliosis in 5xFAD mice. CB1 blockade exacerbated inflammation in this transgenic mouse model of AD. The genetic inactivation of FAAH led to increases in the expression of inflammatory cytokines. At the same time, FAAH-null 5xFAD mice exhibited a behavioral improvement in spatial memory that was independent of the level of anxiety and was not CB1 mediated. Finally, mice lacking FAAH showed diminished soluble amyloid levels, neuritic plaques, and gliosis. These data reinforce the notion of a role for the EC system in neuroinflammation and open new perspectives on the relevance of modulating EC levels in the inflamed brain.
Collapse
Affiliation(s)
- Carmen Vázquez
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Rosa M Tolón
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - M Teresa Grande
- School of Biosciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Marina Caraza
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain; School of Biosciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Marta Moreno
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Erin C Koester
- Department of Pharmacology and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Borja Villaescusa
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Lourdes Ruiz-Valdepeñas
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | | | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA; Department of Cell Biology, The Scripps Research Institute, La Jolla, CA, USA; Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Cecilia J Hillard
- Department of Pharmacology and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Julián Romero
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain; School of Biosciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain.
| |
Collapse
|
41
|
The potential of inhibitors of endocannabinoid metabolism as anxiolytic and antidepressive drugs--A practical view. Eur Neuropsychopharmacol 2015; 25:749-62. [PMID: 25791296 DOI: 10.1016/j.euroneuro.2015.02.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/13/2015] [Accepted: 02/20/2015] [Indexed: 12/15/2022]
Abstract
The endocannabinoid system, comprising cannabinoid CB1 and CB2 receptors, their endogenous ligands anandamide and 2-arachidonoylglyerol, and their synthetic and metabolic enzymes, are involved in many biological processes in the body, ranging from appetite to bone turnover. Compounds inhibiting the breakdown of anandamide and 2-arachidonoylglycerol increase brain levels of these lipids and thus modulate endocannabinoid signalling. In the present review, the preclinical evidence that these enzymes are good targets for development of novel therapies for anxiety and depression are discussed from a practical, rather than mechanistic, point of view. It is concluded that the preclinical data are promising, albeit tempered by problems of tolerance as well as effects upon learning and memory for irreversible monoacylglycerol lipase inhibitors, and limited by a focus upon male rodents alone. Clinical data so far has been restricted to safety studies with inhibitors of anandamide hydrolysis and a hitherto unpublished study on such a compound in elderly patients with major depressive disorders, but under the dose regimes used, they are well tolerated and show no signs of "cannabis-like" behaviours.
Collapse
|
42
|
Burri L, Johnsen L. Krill products: an overview of animal studies. Nutrients 2015; 7:3300-21. [PMID: 25961320 PMCID: PMC4446753 DOI: 10.3390/nu7053300] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 04/29/2015] [Accepted: 05/04/2015] [Indexed: 12/18/2022] Open
Abstract
Many animal studies have been performed with krill oil (KO) and this review aims to summarize their findings and give insight into the mechanism of action of KO. Animal models that have been used in studies with KO include obesity, depression, myocardial infarction, chronic low-grade and ulcerative inflammation and are described in detail. Moreover, studies with KO in the form of krill powder (KP) and krill protein concentrate (KPC) as a mix of lipids and proteins are mentioned and compared to the effects of KO. In addition, differences in tissue uptake of the long-chain omega-3 polyunsaturated fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), when delivered in either phospholipid or triglyceride form, are addressed and the differential impact the delivery form has on gene expression profiles is explained. In our outlook, we try to highlight the potential of KO and KP supplementation in clinical settings and discuss health segments that have a high potential of showing krill product specific health benefits and warrant further clinical investigations.
Collapse
Affiliation(s)
- Lena Burri
- Aker BioMarine Antarctic AS, Fjordalléen 16, NO-0115 Oslo, Norway.
| | - Line Johnsen
- Aker BioMarine Antarctic AS, Fjordalléen 16, NO-0115 Oslo, Norway.
| |
Collapse
|
43
|
Abstract
Corticotropin-releasing hormone (CRH) is a central integrator in the brain of endocrine and behavioral stress responses, whereas activation of the endocannabinoid CB1 receptor suppresses these responses. Although these systems regulate overlapping functions, few studies have investigated whether these systems interact. Here we demonstrate a novel mechanism of CRH-induced anxiety that relies on modulation of endocannabinoids. Specifically, we found that CRH, through activation of the CRH receptor type 1 (CRHR1), evokes a rapid induction of the enzyme fatty acid amide hydrolase (FAAH), which causes a reduction in the endocannabinoid anandamide (AEA), within the amygdala. Similarly, the ability of acute stress to modulate amygdala FAAH and AEA in both rats and mice is also mediated through CRHR1 activation. This interaction occurs specifically in amygdala pyramidal neurons and represents a novel mechanism of endocannabinoid-CRH interactions in regulating amygdala output. Functionally, we found that CRH signaling in the amygdala promotes an anxious phenotype that is prevented by FAAH inhibition. Together, this work suggests that rapid reductions in amygdala AEA signaling following stress may prime the amygdala and facilitate the generation of downstream stress-linked behaviors. Given that endocannabinoid signaling is thought to exert "tonic" regulation on stress and anxiety responses, these data suggest that CRH signaling coordinates a disruption of tonic AEA activity to promote a state of anxiety, which in turn may represent an endogenous mechanism by which stress enhances anxiety. These data suggest that FAAH inhibitors may represent a novel class of anxiolytics that specifically target stress-induced anxiety.
Collapse
|
44
|
Therapeutic potential of inhibitors of endocannabinoid degradation for the treatment of stress-related hyperalgesia in an animal model of chronic pain. Neuropsychopharmacology 2015; 40:488-501. [PMID: 25100669 PMCID: PMC4443964 DOI: 10.1038/npp.2014.198] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/21/2014] [Accepted: 08/01/2014] [Indexed: 11/08/2022]
Abstract
The occurrence of chronic stress, depression, and anxiety can increase nociception in humans and may facilitate the transition from localized to chronic widespread pain. The mechanisms underlying chronic widespread pain are still unknown, hindering the development of effective pharmacological therapies. Here, we exposed C57BL/6J mice to chronic unpredictable stress (CUS) to investigate how persistent stress affects nociception. Next, mice were treated with multiple intramuscular nerve growth factor (NGF) injections, which induced chronic widespread nociception. Thus, combination of CUS and NGF served as a model where psychophysiological impairment coexists with long-lasting hyperalgesia. We found that CUS increased anxiety- and depression-like behavior and enhanced basal nociception in mice. When co-applied with repeated NGF injections, CUS elicited a sustained long-lasting widespread hyperalgesia. In order to evaluate a potential therapeutic strategy for the treatment of chronic pain associated with stress, we hypothesized that the endocannabinoid system (ECS) may represent a target signaling system. We found that URB597, an inhibitor of the anandamide-degrading enzyme fatty acid amide hydrolase (FAAH), and JZL184, an inhibitor of the 2-arachidonoyl glycerol-degrading enzyme monoacylglycerol lipase (MAGL), increased eCB levels in the brain and periphery and were both effective in reducing CUS-induced anxiety measured by the light-dark test and CUS-induced thermal hyperalgesia. Remarkably, the long-lasting widespread hyperalgesia induced by combining CUS and NGF was effectively reduced by URB597, but not by JZL184. Simultaneous inhibition of FAAH and MAGL did not improve the overall therapeutic response. Therefore, our findings indicate that enhancement of anandamide signaling with URB597 is a promising pharmacological approach for the alleviation of chronic widespread nociception in stress-exposed mice, and thus, it could represent a potential treatment strategy for chronic pain associated with neuropsychiatric disorders in humans.
Collapse
|
45
|
Shonesy BC, Bluett RJ, Ramikie TS, Báldi R, Hermanson DJ, Kingsley PJ, Marnett LJ, Winder DG, Colbran RJ, Patel S. Genetic disruption of 2-arachidonoylglycerol synthesis reveals a key role for endocannabinoid signaling in anxiety modulation. Cell Rep 2014; 9:1644-1653. [PMID: 25466252 PMCID: PMC4268380 DOI: 10.1016/j.celrep.2014.11.001] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/09/2014] [Accepted: 10/30/2014] [Indexed: 01/29/2023] Open
Abstract
Endocannabinoid (eCB) signaling has been heavily implicated in the modulation of anxiety and depressive behaviors and emotional learning. However, the role of the most-abundant endocannabinoid 2-arachidonoylglycerol (2-AG) in the physiological regulation of affective behaviors is not well understood. Here, we show that genetic deletion of the 2-AG synthetic enzyme diacylglycerol lipase α (DAGLα) in mice reduces brain, but not circulating, 2-AG levels. DAGLα deletion also results in anxiety-like and sex-specific anhedonic phenotypes associated with impaired activity-dependent eCB retrograde signaling at amygdala glutamatergic synapses. Importantly, acute pharmacological normalization of 2-AG levels reverses both phenotypes of DAGLα-deficient mice. These data suggest 2-AG deficiency could contribute to the pathogenesis of affective disorders and that pharmacological normalization of 2-AG signaling could represent an approach for the treatment of mood and anxiety disorders.
Collapse
Affiliation(s)
- Brian C Shonesy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Rebecca J Bluett
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Teniel S Ramikie
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Rita Báldi
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Daniel J Hermanson
- Departments of Biochemistry, Chemistry, and Pharmacology, A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Philip J Kingsley
- Departments of Biochemistry, Chemistry, and Pharmacology, A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lawrence J Marnett
- Departments of Biochemistry, Chemistry, and Pharmacology, A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Danny G Winder
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt-Kennedy Center for Research on Human Development, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Roger J Colbran
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt-Kennedy Center for Research on Human Development, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sachin Patel
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt-Kennedy Center for Research on Human Development, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
46
|
Rossi S, Motta C, Musella A, Centonze D. The interplay between inflammatory cytokines and the endocannabinoid system in the regulation of synaptic transmission. Neuropharmacology 2014; 96:105-12. [PMID: 25268960 DOI: 10.1016/j.neuropharm.2014.09.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/15/2014] [Accepted: 09/16/2014] [Indexed: 12/14/2022]
Abstract
Excessive glutamate-mediated synaptic transmission and secondary excitotoxicity have been proposed as key determinants of neurodegeneration in many neurological diseases. Soluble mediators of inflammation have recently gained attention owing to their ability to enhance glutamate transmission and affect synaptic sensitivity to neurotransmitters. In the complex crosstalk between soluble immunoactive molecules and synapses, the endocannabinoid system (ECS) plays a central role, exerting an indirect neuroprotective action by inhibiting cytokine-dependent synaptic alterations, and a direct neuroprotective effect by limiting glutamate transmission and excitotoxic damage. On the other hand, the endocannabinoid (eCB)-mediated control of synaptic transmission is altered by proinflammatory cytokines with consequent effects in central nervous system (CNS) disorders. In this review, we summarize the interactions, at the pre- and postsynaptic level, between major inflammatory cytokines and the ECS. In addition, the behavioral and clinical consequences of the modulation of synaptic transmission during neuroinflammation are discussed. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Silvia Rossi
- Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata, 00133 Rome, Italy; Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC), 00143 Rome, Italy
| | - Caterina Motta
- Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata, 00133 Rome, Italy; Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC), 00143 Rome, Italy
| | - Alessandra Musella
- Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata, 00133 Rome, Italy; Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC), 00143 Rome, Italy
| | - Diego Centonze
- Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata, 00133 Rome, Italy; Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC), 00143 Rome, Italy.
| |
Collapse
|
47
|
Central anandamide deficiency predicts stress-induced anxiety: behavioral reversal through endocannabinoid augmentation. Transl Psychiatry 2014; 4:e408. [PMID: 25004388 PMCID: PMC4119220 DOI: 10.1038/tp.2014.53] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/28/2014] [Accepted: 05/21/2014] [Indexed: 01/10/2023] Open
Abstract
Stress is a major risk factor for the development of mood and anxiety disorders; elucidation of novel approaches to mitigate the deleterious effects of stress could have broad clinical applications. Pharmacological augmentation of central endogenous cannabinoid (eCB) signaling may be an effective therapeutic strategy to mitigate the adverse behavioral and physiological consequences of stress. Here we show that acute foot-shock stress induces a transient anxiety state measured 24 h later using the light-dark box assay and novelty-induced hypophagia test. Acute pharmacological inhibition of the anandamide-degrading enzyme, fatty acid amide hydrolase (FAAH), reverses the stress-induced anxiety state in a cannabinoid receptor-dependent manner. FAAH inhibition does not significantly affect anxiety-like behaviors in non-stressed mice. Moreover, whole brain anandamide levels are reduced 24 h after acute foot-shock stress and are negatively correlated with anxiety-like behavioral measures in the light-dark box test. These data indicate that central anandamide levels predict acute stress-induced anxiety, and that reversal of stress-induced anandamide deficiency is a key mechanism subserving the therapeutic effects of FAAH inhibition. These studies provide further support that eCB-augmentation is a viable pharmacological strategy for the treatment of stress-related neuropsychiatric disorders.
Collapse
|
48
|
Laricchiuta D, Musella A, Rossi S, Centonze D. Behavioral and electrophysiological effects of endocannabinoid and dopaminergic systems on salient stimuli. Front Behav Neurosci 2014; 8:183. [PMID: 24904335 PMCID: PMC4032909 DOI: 10.3389/fnbeh.2014.00183] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/04/2014] [Indexed: 01/23/2023] Open
Abstract
Rewarding effects have been related to enhanced dopamine (DA) release in corticolimbic and basal ganglia structures. The DAergic and endocannabinoid interaction in the responses to reward is described. This study investigated the link between endocannabinoid and DAergic transmission in the processes that are related to response to two types of reward, palatable food and novelty. Mice treated with drugs acting on endocannabinoid system (ECS) (URB597, AM251) or DAergic system (haloperidol) were submitted to approach-avoidance conflict tasks with palatable food or novelty. In the same mice, the cannabinoid type-1 (CB1)-mediated GABAergic transmission in medium spiny neurons of the dorsomedial striatum was analyzed. The endocannabinoid potentiation by URB597 magnified approach behavior for reward (food and novelty) and in parallel inhibited dorsostriatal GABAergic neurotransmission. The decreased activity of CB1 receptor by AM251 (alone or with URB597) or of DAergic D2 receptor by haloperidol had inhibitory effects toward the reward and did not permit the inhibition of dorsostriatal GABAergic transmission. When haloperidol was coadministered with URB597, a restoration effect on reward and reward-dependent motor activity was observed, only if the reward was the palatable food. In parallel, the coadministration led to restoring inhibition of CB1-mediated GABAergic transmission. Thus, in the presence of simultaneous ECS activation and inhibition of DAergic system the response to reward appears to be a stimulus-dependent manner.
Collapse
Affiliation(s)
- Daniela Laricchiuta
- IRCCS Fondazione Santa Lucia Rome, Italy ; Dipartimento di Psicologia, Facoltà di Medicina e Psicologia, Università "Sapienza" di Roma Rome, Italy
| | - Alessandra Musella
- IRCCS Fondazione Santa Lucia Rome, Italy ; Dipartimento di Neuroscienze, Università Tor Vergata Rome, Italy
| | - Silvia Rossi
- IRCCS Fondazione Santa Lucia Rome, Italy ; Dipartimento di Neuroscienze, Università Tor Vergata Rome, Italy
| | - Diego Centonze
- IRCCS Fondazione Santa Lucia Rome, Italy ; Dipartimento di Neuroscienze, Università Tor Vergata Rome, Italy
| |
Collapse
|
49
|
Amygdala FAAH and anandamide: mediating protection and recovery from stress. Trends Pharmacol Sci 2013; 34:637-44. [PMID: 24325918 DOI: 10.1016/j.tips.2013.08.008] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/27/2013] [Accepted: 08/29/2013] [Indexed: 11/23/2022]
Abstract
A long-standing literature linking endocannabinoids (ECBs) to stress, fear, and anxiety has led to growing interest in developing novel anxiolytics targeting the ECB system. Following rapid on-demand biosynthesis and degradation upon neuronal activation, the ECB N-arachidonoylethanolamide (anandamide, AEA) is actively degraded by the serine hydrolase enzyme, fatty acid amide hydrolase (FAAH). Exposure to stress rapidly mobilizes FAAH to deplete the signaling pool of AEA and increase neuronal excitability in a key anxiety-mediating region--the basolateral amygdala (BLA). Gene deletion or pharmacological inhibition of FAAH prevents stress-induced reductions in AEA and associated increases in BLA dendritic hypertrophy and anxiety-like behavior. Additionally, inhibition of FAAH facilitates long-term fear extinction and rescues deficient fear extinction in rodent models by enhancing AEA-CB1 (cannabinoid type 1) receptor signaling and synaptic plasticity in the BLA. These preclinical findings propose restoring deficient BLA AEA levels by pharmacologically inhibiting FAAH as a mechanism to therapeutically mitigate the effects of traumatic stress.
Collapse
|
50
|
Hill MN, Patel S. Translational evidence for the involvement of the endocannabinoid system in stress-related psychiatric illnesses. BIOLOGY OF MOOD & ANXIETY DISORDERS 2013; 3:19. [PMID: 24286185 PMCID: PMC3817535 DOI: 10.1186/2045-5380-3-19] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 10/04/2013] [Indexed: 12/30/2022]
Abstract
Accumulating evidence over the past decade has highlighted an important role of the endocannabinoid (eCB) system in the regulation of stress and emotional behavior across divergent species, from rodents to humans. The general findings from this work indicate that the eCB system plays an important role in gating and buffering the stress response, dampening anxiety and regulating mood. Work in rodents has allowed researchers to determine the neural mechanisms mediating this relationship while work in human populations has demonstrated the possible importance of this system in stress-related psychiatric diseases, such as post-traumatic stress disorder, generalized anxiety and major depression. These stress-protective effects of eCB signaling appear to be primarily mediated by their actions within corticolimbic structures, particularly the amygdala and the prefrontal cortex. The aim of this review is to provide an up-to-date discussion of the current level of knowledge in this field, as well as address the current gaps in knowledge and specific areas of research that require attention.
Collapse
Affiliation(s)
- Matthew N Hill
- Hotchkiss Brain Institute, Departments of Cell Biology & Anatomy and Psychiatry, University of Calgary, 3330 Hospital Drive NW, Calgary AB T2N4N1, Canada.
| | | |
Collapse
|