1
|
Gom RC, Wickramarachchi P, George AG, Lightfoot SHM, Newton-Gunderson D, Hill MN, Teskey GC, Colangeli R. Phytocannabinoids restore seizure-induced alterations in emotional behaviour in male rats. Neuropsychopharmacology 2024:10.1038/s41386-024-02005-y. [PMID: 39433952 DOI: 10.1038/s41386-024-02005-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024]
Abstract
Epilepsy often presents with severe emotional comorbidities including anxiety and abnormal fear responses which impose a significant burden on, and reduce, quality of life in people living with the disease. Our lab has recently shown that kindled seizures lead to changes in emotional processing resulting from the downregulation of anandamide signalling within the amygdala. Phytocannabinoids derived from the Cannabis sativa plant have attracted a lot of interest as a new class of drugs with potential anticonvulsant effects. Among the wide number of compounds occurring in Cannabis sativa, Δ9- tetrahydrocannabinol (THC), the one responsible for its main psychoactive effects, and the nonpsychoactive cannabidiol (CBD) have been extensively examined under pre-clinical and clinical contexts to control seizures, however, neither have been assessed in the context of the management of emotional comorbidities associated with seizure activity. We used two behavioural procedures to assess anxiety- and fear-like responding in adult male Long-Evans rats: elevated plus maze and auditory fear conditioning. In agreement with previous reports, we found seizure-induced increases in anxiety- and fear-like responding. These effects were reversed by either CBD (vaporized) or THC (oral). We also found that antagonism of serotonin 1 A receptors prior to CBD exposure prevented its protective effects. Phytocannabinoids offer a novel and reliable opportunity to treat seizure induced comorbid emotional alterations.
Collapse
Affiliation(s)
- Renaud C Gom
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada.
| | - Pasindu Wickramarachchi
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
| | - Antis G George
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
| | - Savannah H M Lightfoot
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Mathison Centre for Mental Health Research and Education; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Dana Newton-Gunderson
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
- Mathison Centre for Mental Health Research and Education; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - G Campbell Teskey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
| | - Roberto Colangeli
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
- Department of Experimental and Clinical Medicine; Università Politecnica delle Marche, 60126, Ancona, Italy
| |
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
|
Reich N, Mannino M, Kotler S. Using caffeine as a chemical means to induce flow states. Neurosci Biobehav Rev 2024; 159:105577. [PMID: 38331128 DOI: 10.1016/j.neubiorev.2024.105577] [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: 10/02/2023] [Revised: 02/01/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
Flow is an intrinsically rewarding state characterised by positive affect and total task absorption. Because cognitive and physical performance are optimal in flow, chemical means to facilitate this state are appealing. Caffeine, a non-selective adenosine receptor antagonist, has been emphasized as a potential flow-inducer. Thus, we review the psychological and biological effects of caffeine that, conceptually, enhance flow. Caffeine may facilitate flow through various effects, including: i) upregulation of dopamine D1/D2 receptor affinity in reward-associated brain areas, leading to greater energetic arousal and 'wanting'; ii) protection of dopaminergic neurons; iii) increases in norepinephrine release and alertness, which offset sleep-deprivation and hypoarousal; iv) heightening of parasympathetic high frequency heart rate variability, resulting in improved cortical stress appraisal, v) modification of striatal endocannabinoid-CB1 receptor-signalling, leading to enhanced stress tolerance; and vi) changes in brain network activity in favour of executive function and flow. We also discuss the application of caffeine to treat attention deficit hyperactivity disorder and caveats. We hope to inspire studies assessing the use of caffeine to induce flow.
Collapse
Affiliation(s)
- Niklas Reich
- Faculty of Health and Medicine, Biomedical & Life Sciences Division, Lancaster University, Lancaster LA1 4YQ, UK; The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK.
| | - Michael Mannino
- Flow Research Collective, USA; Miami Dade College, Miami, FL, USA
| | | |
Collapse
|
4
|
Petrie GN, Balsevich G, Füzesi T, Aukema RJ, Driever WPF, van der Stelt M, Bains JS, Hill MN. Disruption of tonic endocannabinoid signalling triggers cellular, behavioural and neuroendocrine responses consistent with a stress response. Br J Pharmacol 2023; 180:3146-3159. [PMID: 37482931 DOI: 10.1111/bph.16198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/11/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND AND PURPOSE Endocannabinoid (eCB) signalling gates many aspects of the stress response, including the hypothalamic-pituitary-adrenal (HPA) axis. The HPA axis is controlled by corticotropin releasing hormone (CRH) producing neurons in the paraventricular nucleus of the hypothalamus (PVN). Disruption of eCB signalling increases drive to the HPA axis, but the mechanisms subserving this process are poorly understood. EXPERIMENTAL APPROACH Using an array of cellular, endocrine and behavioural readouts associated with activation of CRH neurons in the PVN, we evaluated the contributions of tonic eCB signalling to the generation of a stress response. KEY RESULTS The CB1 receptor antagonist/inverse agonist AM251, neutral antagonist NESS243 and NAPE PLD inhibitor LEI401 all uniformly increased Fos in the PVN, unmasked stress-linked behaviours, such as grooming, and increased circulating CORT, recapitulating the effects of stress. Similar effects were also seen after direct administration of AM251 into the PVN, while optogenetic inhibition of PVN CRH neurons ameliorated stress-like behavioural changes produced by disruption of eCB signalling. CONCLUSIONS AND IMPLICATIONS These data indicate that under resting conditions, constitutive eCB signalling restricts activation of the HPA axis through local regulation of CRH neurons in the PVN.
Collapse
Affiliation(s)
- Gavin N Petrie
- Neuroscience Program, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
| | - Georgia Balsevich
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
| | - Tamás Füzesi
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Robert J Aukema
- Neuroscience Program, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
| | - Wouter P F Driever
- Department of Molecular Physiology, LIC, Leiden University, Leiden, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, LIC, Leiden University, Leiden, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Jaideep S Bains
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
5
|
Hill MN, Haney M, Hillard CJ, Karhson DS, Vecchiarelli HA. The endocannabinoid system as a putative target for the development of novel drugs for the treatment of psychiatric illnesses. Psychol Med 2023; 53:7006-7024. [PMID: 37671673 PMCID: PMC10719691 DOI: 10.1017/s0033291723002465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 09/07/2023]
Abstract
Cannabis is well established to impact affective states, emotion and perceptual processing, primarily through its interactions with the endocannabinoid system. While cannabis use is quite prevalent in many individuals afflicted with psychiatric illnesses, there is considerable controversy as to whether cannabis may worsen these conditions or provide some form of therapeutic benefit. The development of pharmacological agents which interact with components of the endocannabinoid system in more localized and discrete ways then via phytocannabinoids found in cannabis, has allowed the investigation if direct targeting of the endocannabinoid system itself may represent a novel approach to treat psychiatric illness without the potential untoward side effects associated with cannabis. Herein we review the current body of literature regarding the various pharmacological tools that have been developed to target the endocannabinoid system, their impact in preclinical models of psychiatric illness and the recent data emerging of their utilization in clinical trials for psychiatric illnesses, with a specific focus on substance use disorders, trauma-related disorders, and autism. We highlight several candidate drugs which target endocannabinoid function, particularly inhibitors of endocannabinoid metabolism or modulators of cannabinoid receptor signaling, which have emerged as potential candidates for the treatment of psychiatric conditions, particularly substance use disorder, anxiety and trauma-related disorders and autism spectrum disorders. Although there needs to be ongoing clinical work to establish the potential utility of endocannabinoid-based drugs for the treatment of psychiatric illnesses, the current data available is quite promising and shows indications of several potential candidate diseases which may benefit from this approach.
Collapse
Affiliation(s)
- Matthew N. Hill
- Departments of Cell Biology and Anatomy & Psychiatry, Cumming School of Medicine, Hotchkiss Brain Institute and The Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Canada
| | - Margaret Haney
- Department of Psychiatry, New York State Psychiatric Institute and Columbia University Irving Medical Center, New York, USA
| | - Cecilia J. Hillard
- Department of Pharmacology and Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, USA
| | - Debra S. Karhson
- Department of Psychology, University of New Orleans, New Orleans, USA
| | | |
Collapse
|
6
|
Genovese T, Duranti A, Monaco F, Siracusa R, Fusco R, Impellizzeri D, D’Amico R, Cordaro M, Cuzzocrea S, Di Paola R. Inhibition of Fatty Acid Amide Hydrolase (FAAH) Regulates NF-kb Pathways Reducing Bleomycin-Induced Chronic Lung Inflammation and Pulmonary Fibrosis. Int J Mol Sci 2023; 24:10125. [PMID: 37373275 PMCID: PMC10298572 DOI: 10.3390/ijms241210125] [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: 04/26/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
The deadly interstitial lung condition known as idiopathic pulmonary fibrosis (IPF) worsens over time and for no apparent reason. The traditional therapy approaches for IPF, which include corticosteroids and immunomodulatory drugs, are often ineffective and can have noticeable side effects. The endocannabinoids are hydrolyzed by a membrane protein called fatty acid amide hydrolase (FAAH). Increasing endogenous levels of endocannabinoid by pharmacologically inhibiting FAAH results in numerous analgesic advantages in a variety of experimental models for pre-clinical pain and inflammation. In our study, we mimicked IPF by administering intratracheal bleomycin, and we administered oral URB878 at a dose of 5 mg/kg. The histological changes, cell infiltration, pro-inflammatory cytokine production, inflammation, and nitrosative stress caused by bleomycin were all reduced by URB878. Our data clearly demonstrate for the first time that the inhibition of FAAH activity was able to counteract not only the histological alteration bleomycin-induced but also the cascade of related inflammatory events.
Collapse
Affiliation(s)
- Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento, 6, 61029 Urbino, Italy
| | - Francesco Monaco
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98166 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| |
Collapse
|
7
|
Santoso AD, De Ridder D. Fatty Acid Amide Hydrolase: An Integrative Clinical Perspective. Cannabis Cannabinoid Res 2023; 8:56-76. [PMID: 35900294 DOI: 10.1089/can.2021.0237] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Introduction: Fatty acid amide hydrolase (FAAH) is one of the main terminating enzymes of the endocannabinoid system (ECS). Since being discovered in 1996, the modulation of FAAH has been viewed as a compelling alternative strategy to obtain the beneficial effect of the ECS. With a considerable amount of FAAH-related publication over time, the next step would be to comprehend the proximity of this evidence for clinical application. Objective: This review intends to highlight the rationale of FAAH modulation and provide the latest evidence from clinical studies. Methods: Publication searches were conducted to gather information focused on FAAH-related clinical evidence with an extension to the experimental research to understand the biological plausibility. The subtopics were selected to be multidisciplinary to offer more perspective on the current state of the arts. Discussion: Experimental and clinical studies have demonstrated that FAAH was highly expressed not only in the central nervous system but also in the peripheral tissues. As the key regulator of endocannabinoid signaling, it would appear that FAAH plays a role in the modulation of mood and emotional response, reward system, pain perception, energy metabolism and appetite regulation, inflammation, and other biological processes. Genetic variants may be associated with some conditions such as substance/alcohol use disorders, obesity, and eating disorder. The advancement of functional neuroimaging has enabled the evaluation of the neurochemistry of FAAH in brain tissues and this can be incorporated into clinical trials. Intriguingly, the application of FAAH inhibitors in clinical trials seems to provide less striking results in comparison with the animal models, although some potential still can be seen. Conclusion: Modulation of FAAH has an immense potential to be a new therapeutic candidate for several disorders. Further exploration, however, is still needed to ensure who is the best candidate for the treatment strategy.
Collapse
Affiliation(s)
- Anugrah D Santoso
- Laboratory of Experimental Urology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Urology, Faculty of Medicine Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Dirk De Ridder
- Laboratory of Experimental Urology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Zhong H, Rong J, Yang Y, Liang M, Li Y, Zhou R. Neonatal inflammation via persistent TGF-β1 downregulation decreases GABA AR expression in basolateral amygdala leading to the imbalance of the local excitation-inhibition circuits and anxiety-like phenotype in adult mice. Neurobiol Dis 2022; 169:105745. [PMID: 35513229 DOI: 10.1016/j.nbd.2022.105745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/29/2022] Open
Abstract
Neonatal inflammation can increase the risk of anxiety disorder in adulthood. The balance between glutamatergic excitatory and GABAergic inhibitory transmissions in the basolateral amygdala (BLA) plays a vital role in controlling anxiety state. Based on the reports that early-life inflammation had adverse effects on GABAergic system, the aim of this study was to investigate whether and how neonatal inflammation affects excitatory-inhibitory circuits in the BLA resulting in anxiety disorder. Neonatal mice received a daily subcutaneous injection of lipopolysaccharide (LPS, 50 μg/kg) or saline on postnatal days 3-5. LPS-treated mice developed anxiety behaviors accompanied by the hyperactivity of adrenal axis in adulthood. Electrophysiological study revealed the increase of postsynaptic neuronal excitability in the cortical-BLA excitatory synapses of LPS mice which could be recovered by bath-application of GABAAR agonist suggesting the impairment of GABAergic system in LPS mice. Compared with controls, GABAARα2 subunit expression and density of GABA-evoked current in BLA principal neurons were reduced in LPS mice. Additionally, neonatal LPS treatment resulted in the down-regulation of transforming growth factor-beta 1 (TGF-β1) expression and PKC signaling pathway in the adult BLA. The local TGF-β1 overexpression in the BLA improved GABAARα2 expression via up-regulating the activity of PKC signaling, which corrected GABAAR-mediated inhibition leading to the abolishment of anxiety-like change in adrenal axis regulation and behaviors in LPS mice. These data suggest the persistent TGF-β1deficit induces the down-regulation of GABAARα2 expression and subsequent disruption of the excitation-inhibition balance in the BLA circuits, which is the important mechanisms of neonatal inflammation-induced anxiety disorder.
Collapse
Affiliation(s)
- Haiquan Zhong
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Jing Rong
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Yang Yang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Min Liang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Yingchun Li
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Rong Zhou
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
9
|
Genovese T, Duranti A, D’Amico R, Fusco R, Impellizzeri D, Peritore AF, Crupi R, Gugliandolo E, Cuzzocrea S, Di Paola R, Siracusa R, Cordaro M. Fatty Acid Amide Hydrolase (FAAH) Inhibition Plays a Key Role in Counteracting Acute Lung Injury. Int J Mol Sci 2022; 23:2781. [PMID: 35269926 PMCID: PMC8910911 DOI: 10.3390/ijms23052781] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/26/2022] [Accepted: 03/01/2022] [Indexed: 02/01/2023] Open
Abstract
Acute lung injury (ALI) is a group of lung illnesses characterized by severe inflammation, with no treatment. The fatty acid amide hydrolase (FAAH) enzyme is an integral membrane protein responsible for the hydrolysis of the main endocannabinoids, such as anandamide (AEA). In pre-clinical pain and inflammation models, increasing the endogenous levels of AEA and other bioactive fatty acid amides (FAAs) via genetic deletion or the pharmacological inhibition of FAAH produces many analgesic benefits in several different experimental models. To date, nobody has investigated the role of FAAH inhibition on an ALI mouse model. Mice were subjected to a carrageenan injection and treated orally 1 h after with the FAAH inhibitor URB878 dissolved in a vehicle consisting of 10% PEG-400, 10% Tween-80 and 80% saline at different doses: The inhibition of FAAH activity was able to counteract not only the CAR-induced histological alteration, but also the cascade of related inflammatory events. URB878 clears the way for further studies based on FAAH inhibition in acute lung pathologies.
Collapse
Affiliation(s)
- Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino, Carlo Bo Piazza del Rinascimento 6, 61029 Urbino, Italy;
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
| | - Roberta Fusco
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (R.C.); (E.G.)
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (R.C.); (E.G.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (R.C.); (E.G.)
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
| | - Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| |
Collapse
|
10
|
Colangeli R, Teskey GC, Di Giovanni G. Endocannabinoid-serotonin systems interaction in health and disease. PROGRESS IN BRAIN RESEARCH 2021; 259:83-134. [PMID: 33541682 DOI: 10.1016/bs.pbr.2021.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endocannabinoid (eCB) and serotonin (5-HT) neuromodulatory systems work both independently and together to finely orchestrate neuronal activity throughout the brain to strongly sculpt behavioral functions. Surprising parallelism between the behavioral effects of 5-HT and eCB activity has been widely reported, including the regulation of emotional states, stress homeostasis, cognitive functions, food intake and sleep. The distribution pattern of the 5-HT system and the eCB molecular elements in the brain display a strong overlap and several studies report a functional interplay and even a tight interdependence between eCB/5-HT signaling. In this review, we examine the available evidence of the interaction between the eCB and 5-HT systems. We first introduce the eCB system, then we describe the eCB/5-HT crosstalk at the neuronal and synaptic levels. Finally, we explore the potential eCB/5-HT interaction at the behavioral level with the implication for psychiatric and neurological disorders. The precise elucidation of how this neuromodulatory interaction dynamically regulates biological functions may lead to the development of more targeted therapeutic strategies for the treatment of depressive and anxiety disorders, psychosis and epilepsy.
Collapse
Affiliation(s)
- Roberto Colangeli
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| | - G Campbell Teskey
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Giuseppe Di Giovanni
- Laboratory of Neurophysiology, Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta; Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
11
|
Ahmad H, Rauf K, Zada W, McCarthy M, Abbas G, Anwar F, Shah AJ. Kaempferol Facilitated Extinction Learning in Contextual Fear Conditioned Rats via Inhibition of Fatty-Acid Amide Hydrolase. Molecules 2020; 25:molecules25204683. [PMID: 33066366 PMCID: PMC7587337 DOI: 10.3390/molecules25204683] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Fear, stress, and anxiety-like behaviors originate from traumatic events in life. Stress response is managed by endocannabinoids in the body by limiting the uncontrolled retrieval of aversive memories. Pharmacotherapy-modulating endocannabinoids, especially anandamide, presents a promising tool for treating anxiety disorders. Here, we investigated the effect of kaempferol, a flavonoid, in the extinction of fear related memories and associated anxiety-like behavior. Methods: The ability of kaempferol to inhibit fatty-acid amide hydrolase (FAAH, the enzyme that catabolizes anandamide) was assessed in vitro using an enzyme-linked immunosorbent assay (ELISA) kit. For animal studies (in vivo), the extinction learning was evaluated using contextual fear conditioning (CFC, a behavioral paradigm based on ability to learn and remember aversive stimuli). Furthermore, an elevated plus-maze (EPM) model was used for measuring anxiety-like behavior, while serum corticosterone served as a biochemical indicator of anxiety. Lastly, the interaction of kaempferol with FAAH enzyme was also assessed in silico (computational study). Results: Our data showed that kaempferol inhibited the FAAH enzyme with an IC50 value of 1 µM. In CFC, it reduced freezing behavior in rats. EPM data demonstrated anxiolytic activity as exhibited by enhanced number of entries and time spent in the open arm. No change in blood corticosterone levels was noted. Our computational study showed that Kaempferol interacted with the catalytic amino acids (SER241, PHE192, PHE381, and THR377) of FAAH enzyme Conclusion: Our study demonstrate that kaempferol facilitated the extinction of aversive memories along with a reduction of anxiety. The effect is mediated through the augmentation of endocannabinoids via the inhibition of FAAH enzyme.
Collapse
Affiliation(s)
- Hammad Ahmad
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Khyber Pakhtunkhwa 22060, Pakistan; (H.A.); (K.R.); (W.Z.)
| | - Khalid Rauf
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Khyber Pakhtunkhwa 22060, Pakistan; (H.A.); (K.R.); (W.Z.)
| | - Wahid Zada
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Khyber Pakhtunkhwa 22060, Pakistan; (H.A.); (K.R.); (W.Z.)
| | - Margaret McCarthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Ghulam Abbas
- Department of Pharmacology, Faculty of Pharmacy, Ziauddin University, Karachi 75000, Pakistan;
| | - Fareeha Anwar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore 54000, Pakistan;
| | - Abdul Jabbar Shah
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Khyber Pakhtunkhwa 22060, Pakistan; (H.A.); (K.R.); (W.Z.)
- Correspondence: ; Tel.: +(92)992-383591-6
| |
Collapse
|
12
|
Anandamide Signaling Augmentation Rescues Amygdala Synaptic Function and Comorbid Emotional Alterations in a Model of Epilepsy. J Neurosci 2020; 40:6068-6081. [PMID: 32601243 DOI: 10.1523/jneurosci.0068-20.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 05/14/2020] [Accepted: 06/21/2020] [Indexed: 12/31/2022] Open
Abstract
Epilepsy is often associated with emotional disturbances and the endocannabinoid (eCB) system tunes synaptic transmission in brain regions regulating emotional behavior. Thus, persistent alteration of eCB signaling after repeated seizures may contribute to the development of epilepsy-related emotional disorders. Here we report that repeatedly eliciting seizures (kindling) in the amygdala caused a long-term increase in anxiety and impaired fear memory retention, which was paralleled by an imbalance in GABA/glutamate presynaptic activity and alteration of synaptic plasticity in the basolateral amygdala (BLA), in male rats. Anandamide (AEA) content was downregulated after repeated seizures, and pharmacological enhancement of AEA signaling rescued seizure-induced anxiety by restoring the tonic control of the eCB signaling over glutamatergic transmission. Moreover, AEA signaling augmentation also rescued the seizure-induced alterations of fear memory by restoring the phasic control of eCB signaling over GABAergic activity and plasticity in the BLA. These results indicate that modulation of AEA signaling represents a potential and promising target for the treatment of comorbid emotional dysfunction associated with epilepsy.SIGNIFICANCE STATEMENT Epilepsy is a heterogeneous neurologic disorder commonly associated with comorbid emotional alterations. However, the management of epilepsy is usually restricted to the control of seizures. The endocannabinoid (eCB) system, particularly anandamide (AEA) signaling, controls neuronal excitability and seizure expression and regulates emotional behavior. We found that repeated seizures cause an allostatic maladaptation of AEA signaling in the amygdala that drives emotional alterations. Boosting AEA signaling through inhibition of its degradative enzyme, fatty acid amide hydrolase (FAAH), restored both synaptic and behavioral alterations. FAAH inhibitors dampen seizure activity in animal models and are used in clinical studies to treat the negative consequences associated with stress. Thereby, they are accessible and can be clinically evaluated to treat both seizures and comorbid conditions associated with epilepsy.
Collapse
|
13
|
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: 101] [Impact Index Per Article: 25.3] [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
|
14
|
Sbarski B, Akirav I. Cannabinoids as therapeutics for PTSD. Pharmacol Ther 2020; 211:107551. [PMID: 32311373 DOI: 10.1016/j.pharmthera.2020.107551] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 03/08/2020] [Indexed: 02/09/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a complex disorder that involves dysregulation of multiple neurobiological systems. The traumatic stressor plays a causal role in producing psychological dysfunction and the pattern of findings suggests that the hypothalamic-pituitary-adrenal (HPA) axis, which is instrumental for stress adaptation, is critically dysfunctional in PTSD. Given the lack of understanding of the basic mechanisms and underlying pathways that cause the disorder and its heterogeneity, PTSD poses challenges for treatment. Targeting the endocannabinoid (ECB) system to treat mental disorders, and PTSD in particular, has been the focus of research and interest in recent years. The ECB system modulates multiple functions, and drugs enhancing ECB signaling have shown promise as potential therapeutic agents in stress effects and other psychiatric and medical conditions. In this review, we focus on the interaction between the ECB-HPA systems in animal models for PTSD and in patients with PTSD. We summarize evidence supporting the use of cannabinoids in preventing and treating PTSD in preclinical and clinical studies. As the HPA system plays a key role in the mediation of the stress response and the pathophysiology of PTSD, we describe preclinical studies suggesting that enhancing ECB signaling is consistent with decreasing PTSD symptoms and dysfunction of the HPA axis. Overall, we suggest that a pharmacological treatment targeted at one system (e.g., HPA) may not be very effective because of the heterogeneity of the disorder. There are abnormalities across different neurotransmitter systems in the pathophysiology of PTSD and none of these systems function uniformly among all patients with PTSD. Hence, conceptually, enhancing ECB signaling may be a more effective avenue for pharmacological treatment.
Collapse
Affiliation(s)
- Brenda Sbarski
- School of Psychological Sciences, Integrated Brain and Behavior Research Center, University of Haifa, Haifa 3498838, Israel
| | - Irit Akirav
- School of Psychological Sciences, Integrated Brain and Behavior Research Center, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
15
|
Mayo LM, Asratian A, Lindé J, Morena M, Haataja R, Hammar V, Augier G, Hill MN, Heilig M. Elevated Anandamide, Enhanced Recall of Fear Extinction, and Attenuated Stress Responses Following Inhibition of Fatty Acid Amide Hydrolase: A Randomized, Controlled Experimental Medicine Trial. Biol Psychiatry 2020; 87:538-547. [PMID: 31590924 DOI: 10.1016/j.biopsych.2019.07.034] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/16/2019] [Accepted: 07/30/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Posttraumatic stress disorder, an area of large unmet medical needs, is characterized by persistence of fear memories and maladaptive stress responses. In rodents, elevation of the endocannabinoid anandamide due to inhibition of fatty acid amide hydrolase (FAAH) facilitates fear extinction and protects against the anxiogenic effects of stress. We recently reported that elevated anandamide levels in people homozygous for a loss-of-function FAAH mutation are associated with a similar phenotype, suggesting a translational validity of the preclinical findings. METHODS In this double-blind, placebo-controlled experimental medicine study, healthy adults were randomized to an FAAH inhibitor (PF-04457845, 4 mg orally, once daily; n = 16) or placebo (n = 29) for 10 days. On days 9 and 10, participants completed a task battery assessing psychophysiological indices of fear learning, stress reactivity, and stress-induced affective responses. RESULTS FAAH inhibition produced a 10-fold increase in baseline anandamide. This was associated with potentiated recall of fear extinction memory when tested 24 hours after extinction training. FAAH inhibition also attenuated autonomic stress reactivity, assessed via electrodermal activity, and protected against stress-induced negative affect, measured via facial electromyography. CONCLUSIONS Our data provide preliminary human evidence that FAAH inhibition can improve the recall of fear extinction memories and attenuate the anxiogenic effects of stress, in a direct translation of rodent findings. The beneficial effects of FAAH inhibition on fear extinction, as well as stress- and affect-related behaviors, provide a strong rationale for developing this drug class as a treatment for posttraumatic stress disorder.
Collapse
Affiliation(s)
- Leah M Mayo
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Anna Asratian
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Johan Lindé
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Maria Morena
- Hotchkiss Brain Institute and Mathison Centre for Mental Health Research and Education, Cummings Scool of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Roosa Haataja
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Valter Hammar
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Gaëlle Augier
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Matthew N Hill
- Hotchkiss Brain Institute and Mathison Centre for Mental Health Research and Education, Cummings Scool of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Markus Heilig
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
16
|
Remote CB1 receptor antagonist administration reveals multiple sites of tonic and phasic endocannabinoid neuroendocrine regulation. Psychoneuroendocrinology 2020; 113:104549. [PMID: 31884322 PMCID: PMC7566018 DOI: 10.1016/j.psyneuen.2019.104549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/21/2019] [Accepted: 12/13/2019] [Indexed: 11/23/2022]
Abstract
Endogenous cannabinoids (endocannabinoids, eCB) are expressed throughout the body and contribute to regulation of the hypothalamo-pituitary-adrenal (HPA) axis and general stress reactivity. This study assessed the contributions of CB1 receptors (CB1R) in the modulation of basal and stress-induced neural and HPA axis activities. Catheterized adult male rats were placed in chambers to acclimate overnight, with their catheters connected and exteriorized from the chambers for relatively stress-free remote injections. The next morning, the CB1R antagonist AM251 (1 or 2 mg/kg) or vehicle was administered, and 30 min later, rats were exposed to loud noise stress (30 min) or no noise (basal condition). Blood, brains, pituitary and adrenal glands were collected immediately after the procedures for analysis of c-fos and CB1R mRNAs, corticosterone (CORT) and adrenocorticotropin hormone (ACTH) plasma levels. Basally, CB1R antagonism induced c-fos mRNA in the basolateral amygdala (BLA) and auditory cortex (AUD) and elevated plasma CORT, indicating disruption of eCB-mediated constitutive inhibition of activity. CB1R blockade also potentiated stress-induced hormone levels and c-fos mRNA in several regions such as the bed nucleus of the stria terminalis (BST), lateral septum (LS), and basolateral amygdala (BLA) and the paraventricular nucleus of the hypothalamus (PVN). CB1R mRNA was detected in all central tissues investigated, and the adrenal cortex, but at very low levels in the anterior pituitary gland. Interestingly, CB1R mRNA was rapidly and bidirectionally regulated in response to stress and/or antagonist treatment in some regions. eCBs therefore modulate the HPA axis by regulating both constitutive and activity-dependent inhibition at multiple levels.
Collapse
|
17
|
Bedse G, Centanni SW, Winder DG, Patel S. Endocannabinoid Signaling in the Central Amygdala and Bed Nucleus of the Stria Terminalis: Implications for the Pathophysiology and Treatment of Alcohol Use Disorder. Alcohol Clin Exp Res 2019; 43:2014-2027. [PMID: 31373708 PMCID: PMC6779484 DOI: 10.1111/acer.14159] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/18/2019] [Indexed: 12/23/2022]
Abstract
High rates of relapse are a chronic and debilitating obstacle to effective treatment of alcohol use disorder (AUD); however, no effective treatments are available to treat symptoms induced by protracted abstinence. In the first part of this 2-part review series, we examine the literature supporting the effects of alcohol exposure within the extended amygdala (EA) neural circuitry. In Part 2, we focus on a potential way to combat negative affect associated with AUD, by exploring the therapeutic potential of the endogenous cannabinoid (eCB) system. The eCB system is a potent modulator of neural activity in the brain, and its ability to mitigate stress and negative affect has long been an area of interest for developing novel therapeutics. This review details the recent advances in our understanding of eCB signaling in 2 key regions of the EA, the central nucleus of the amygdala and the bed nucleus of the stria terminalis (BNST), and their role in regulating negative affect. Despite an established role for EA eCB signaling in reducing negative affect, few studies have examined the potential for eCB-based therapies to treat AUD-associated negative affect. In this review, we present an overview of studies focusing on eCB signaling in EA and cannabinoid modulation on EA synaptic activity. We further discuss studies suggesting dysregulation of eCB signaling in models of AUD and propose that pharmacological augmentation of eCB could be a novel approach to treat aspects of AUD. Lastly, future directions are proposed to advance our understanding of the relationship between AUD-associated negative affect and the EA eCB system that could yield new pharmacotherapies targeting negative affective symptoms associated with AUD.
Collapse
Affiliation(s)
- Gaurav Bedse
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, the, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
| | - Danny G. Winder
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, the, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Sachin Patel
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, the, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| |
Collapse
|
18
|
Ney LJ, Matthews A, Bruno R, Felmingham KL. Cannabinoid interventions for PTSD: Where to next? Prog Neuropsychopharmacol Biol Psychiatry 2019; 93:124-140. [PMID: 30946942 DOI: 10.1016/j.pnpbp.2019.03.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/20/2019] [Accepted: 03/29/2019] [Indexed: 01/18/2023]
Abstract
Cannabinoids are a promising method for pharmacological treatment of post-traumatic stress disorder (PTSD). Despite considerable research devoted to the effect of cannabinoid modulation on PTSD symptomology, there is not a currently agreed way by which the cannabinoid system should be targeted in humans. In this review, we present an overview of recent research identifying neurological pathways by which different cannabinoid-based treatments may exert their effects on PTSD symptomology. We evaluate the strengths and weaknesses of each of these different approaches, including recent challenges presented to favourable options such as fatty acid amide hydrolase (FAAH) inhibitors. This article makes the strengths and challenges of different potential cannabinoid treatments accessible to psychological researchers interested in cannabinoid therapeutics and aims to aid selection of appropriate tools for future clinical trials.
Collapse
Affiliation(s)
- Luke J Ney
- School of Psychology, University of Tasmania, Australia.
| | | | | | - Kim L Felmingham
- School of Psychological Sciences, University of Melbourne, Australia
| |
Collapse
|
19
|
Wirz L, Reuter M, Felten A, Schwabe L. An endocannabinoid receptor polymorphism modulates affective processing under stress. Soc Cogn Affect Neurosci 2019; 13:1177-1189. [PMID: 30239920 PMCID: PMC6234318 DOI: 10.1093/scan/nsy083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 09/17/2018] [Indexed: 02/04/2023] Open
Abstract
Stress has a critical impact on affective and cognitive processing. Based on rodent data suggesting that endocannabinoid signaling via CB1 receptors serves as an emotional buffer, we hypothesized that a common variant of the gene coding for the CB1 receptor modulates affective processing under stress (CNR1; rs1049353 A vs G allele). Therefore, 139 participants, genotyped for this polymorphism, underwent a stress or control manipulation before they viewed emotionally neutral and negative pictures in a magnetic resonance imaging scanner. The ventromedial prefrontal cortex, known for its crucial role in emotion regulation, was significantly more activated in AA/AG vs GG genotype carriers when viewing negative pictures after stress. Under no-stress conditions, AA/AG genotype carriers showed enhanced crosstalk between the ventrolateral prefrontal cortex and the amygdala. We further assessed participants' 24 h-delayed memory for the presented pictures and found that memory performance correlated with amygdala and hippocampus activity and connectivity in stressed carriers of the AA/AG but not the GG genotype. These findings underline the modulatory role of the endocannabinoid system in stress effects on emotion and cognition and provide insights into the neural mechanisms that may contribute to the suggested protective effect of the AA/AG genotype of the CB1 receptor polymorphism against stress-related psychopathologies.
Collapse
Affiliation(s)
- Lisa Wirz
- Department of Cognitive Psychology, University of Hamburg, Hamburg Germany
| | - Martin Reuter
- Department of Differential and Biological Psychology, University of Bonn, Bonn, Germany
| | - Andrea Felten
- Department of Differential and Biological Psychology, University of Bonn, Bonn, Germany
| | - Lars Schwabe
- Department of Cognitive Psychology, University of Hamburg, Hamburg Germany
| |
Collapse
|
20
|
Morena M, Aukema RJ, Leitl KD, Rashid AJ, Vecchiarelli HA, Josselyn SA, Hill MN. Upregulation of Anandamide Hydrolysis in the Basolateral Complex of Amygdala Reduces Fear Memory Expression and Indices of Stress and Anxiety. J Neurosci 2019; 39:1275-1292. [PMID: 30573646 PMCID: PMC6381235 DOI: 10.1523/jneurosci.2251-18.2018] [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: 08/31/2018] [Revised: 12/09/2018] [Accepted: 12/14/2018] [Indexed: 11/21/2022] Open
Abstract
Increased anandamide (AEA) signaling through inhibition of its catabolic enzyme fatty acid amide hydrolase (FAAH) in the basolateral complex of amygdala (BLA) is thought to buffer against the effects of stress and reduces behavioral signs of anxiety and fear. However, examining the role of AEA signaling in stress, anxiety, and fear through pharmacological depletion has been challenging due to the redundant complexity of its biosynthesis and the lack of a pharmacological synthesis inhibitor. We developed a herpes simplex viral vector to rapidly yet transiently overexpress FAAH specifically within the BLA to assess the impact of suppressing AEA signaling on stress, fear, and anxiety in male rats. Surprisingly, FAAH overexpression in BLA dampened stress-induced corticosterone release, reduced anxiety-like behaviors, and decreased conditioned fear expression. Interestingly, depleting AEA signaling in the BLA did not prevent fear conditioning itself or fear reinstatement. These effects were specific to the overexpression of FAAH because they were reversed by intra-BLA administration of an FAAH inhibitor. Moreover, the fear-suppressive effects of FAAH overexpression were also mitigated by intra-BLA administration of a low dose of a GABAA receptor antagonist, but not an NMDA/AMPA/kainate receptor antagonist, suggesting that they were mediated by an increase in GABAergic neurotransmission. Our data suggest that a permissive AEA tone within the BLA might gate GABA release and that loss of this tone through elevated AEA hydrolysis increases inhibition in the BLA, which in turn reduces stress, anxiety, and fear. These data provide new insights on the mechanisms by which amygdalar endocannabinoid signaling regulates emotional behavior.SIGNIFICANCE STATEMENT Amygdala endocannabinoid signaling is involved in the regulation of stress, anxiety, and fear. Our data indicate that viral-mediated augmentation of anandamide hydrolysis within the basolateral amygdala reduces behavioral indices of stress, anxiety, and conditioned fear expression. These same effects have been previously documented with inhibition of anandamide hydrolysis in the same brain region. Our results indicate that the ability of anandamide signaling to regulate emotional behavior is nonlinear and may involve actions at distinct neuronal populations, which could be influenced by the basal level of anandamide. Modulation of anandamide signaling is a current clinical therapeutic target for stress-related psychiatric illnesses, so these data underscore the importance of fully understanding the mechanisms by which anandamide signaling regulates amygdala-dependent changes in emotionality.
Collapse
Affiliation(s)
- Maria Morena
- Hotchkiss Brain Institute,
- Mathison Centre for Mental Health Research
- Departments of Cell Biology and Anatomy and Psychiatry
| | - Robert J Aukema
- Hotchkiss Brain Institute
- Mathison Centre for Mental Health Research
- Neuroscicence Program, Cumming School of Medicine, University of Calgary, T2N 4N1 Calgary, Alberta, Canada, and
| | - Kira D Leitl
- Hotchkiss Brain Institute
- Mathison Centre for Mental Health Research
| | - Asim J Rashid
- Hospital for Sick Children and Departments of Psychology and Physiology, University of Toronto, M5G 1X8 Toronto, Ontario, Canada
| | - Haley A Vecchiarelli
- Hotchkiss Brain Institute
- Mathison Centre for Mental Health Research
- Neuroscicence Program, Cumming School of Medicine, University of Calgary, T2N 4N1 Calgary, Alberta, Canada, and
| | - Sheena A Josselyn
- Hospital for Sick Children and Departments of Psychology and Physiology, University of Toronto, M5G 1X8 Toronto, Ontario, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute,
- Mathison Centre for Mental Health Research
- Departments of Cell Biology and Anatomy and Psychiatry
| |
Collapse
|
21
|
Stopponi S, Fotio Y, Domi A, Borruto AM, Natividad L, Roberto M, Ciccocioppo R, Cannella N. Inhibition of fatty acid amide hydrolase in the central amygdala alleviates co-morbid expression of innate anxiety and excessive alcohol intake. Addict Biol 2018; 23:1223-1232. [PMID: 29071769 DOI: 10.1111/adb.12573] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/15/2017] [Indexed: 01/18/2023]
Abstract
Fatty acid amide hydrolase (FAAH) is an enzyme that prominently degrades the major endocannabinoid N-arachidonoylethanolamine (anandamide). Inhibition of this enzyme leads to increased anandamide levels in brain regions that modulate stress and anxiety. Recently, we found that genetically selected Marchigian Sardinian alcohol-preferring (msP) rats display hyperactive FAAH in amygdalar regions that was associated with increased stress sensitivity and a hyper-anxious phenotype. Our previous work has also demonstrated that msPs display an innate preference for and excessive consumption of alcohol, potentially reflecting a form of self-medication to gain relief from hyper-anxious states. Here, we expand on our previous work by microinjecting the selective FAAH inhibitor URB597 (vehicle, 0.03, 0.1 and 1.0 μg per rat) into the central amygdala (CeA) and basolateral amygdala in msP versus non-selected Wistar rats to evaluate the effects of localized FAAH inhibition on operant alcohol self-administration and restraint-induced anxiety using the elevated plus maze. Intra-CeA URB597 significantly reduced alcohol self-administration in msP but not in Wistar rats. Intra-basolateral amygdala URB597 also attenuated alcohol drinking in msPs, although the effect was less pronounced relative to CeA treatment. In contrast, control experiments administering URB597 into the ventral tegmental area produced no genotypic differences in drinking. We also found that URB597 treatment in the CeA significantly reduced the anxiogenic effects of restraint stress in msPs, although no effects were detected in Wistars. Dysregulation of FAAH regulated systems in the major output region of the amygdala may drive the propensity for co-morbid expression of anxiety and excessive alcohol use.
Collapse
Affiliation(s)
- Serena Stopponi
- School of Pharmacy, Pharmacology Unit; University of Camerino; Camerino Italy
| | - Yannick Fotio
- School of Pharmacy, Pharmacology Unit; University of Camerino; Camerino Italy
| | - Ana Domi
- School of Pharmacy, Pharmacology Unit; University of Camerino; Camerino Italy
| | - Anna Maria Borruto
- School of Pharmacy, Pharmacology Unit; University of Camerino; Camerino Italy
| | - Luis Natividad
- Department of Neuroscience; The Scripps Research Institute; La Jolla CA USA
| | - Marisa Roberto
- Department of Neuroscience; The Scripps Research Institute; La Jolla CA USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit; University of Camerino; Camerino Italy
| | - Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit; University of Camerino; Camerino Italy
| |
Collapse
|
22
|
Gallelli CA, Calcagnini S, Romano A, Koczwara JB, de Ceglia M, Dante D, Villani R, Giudetti AM, Cassano T, Gaetani S. Modulation of the Oxidative Stress and Lipid Peroxidation by Endocannabinoids and Their Lipid Analogues. Antioxidants (Basel) 2018; 7:E93. [PMID: 30021985 PMCID: PMC6070960 DOI: 10.3390/antiox7070093] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/10/2018] [Accepted: 07/13/2018] [Indexed: 02/06/2023] Open
Abstract
Growing evidence supports the pivotal role played by oxidative stress in tissue injury development, thus resulting in several pathologies including cardiovascular, renal, neuropsychiatric, and neurodegenerative disorders, all characterized by an altered oxidative status. Reactive oxygen and nitrogen species and lipid peroxidation-derived reactive aldehydes including acrolein, malondialdehyde, and 4-hydroxy-2-nonenal, among others, are the main responsible for cellular and tissue damages occurring in redox-dependent processes. In this scenario, a link between the endocannabinoid system (ECS) and redox homeostasis impairment appears to be crucial. Anandamide and 2-arachidonoylglycerol, the best characterized endocannabinoids, are able to modulate the activity of several antioxidant enzymes through targeting the cannabinoid receptors type 1 and 2 as well as additional receptors such as the transient receptor potential vanilloid 1, the peroxisome proliferator-activated receptor alpha, and the orphan G protein-coupled receptors 18 and 55. Moreover, the endocannabinoids lipid analogues N-acylethanolamines showed to protect cell damage and death from reactive aldehydes-induced oxidative stress by restoring the intracellular oxidants-antioxidants balance. In this review, we will provide a better understanding of the main mechanisms triggered by the cross-talk between the oxidative stress and the ECS, focusing also on the enzymatic and non-enzymatic antioxidants as scavengers of reactive aldehydes and their toxic bioactive adducts.
Collapse
Affiliation(s)
- Cristina Anna Gallelli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Silvio Calcagnini
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Justyna Barbara Koczwara
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Marialuisa de Ceglia
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Donatella Dante
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Rosanna Villani
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Department of Medical and Surgical Sciences, Institute of Internal Medicine, University of Foggia, 71122 Foggia, Italy.
| | - Anna Maria Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, Via Luigi Pinto, c/o Ospedali Riuniti, 71122 Foggia, Italy.
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
23
|
Reynoso-Moreno I, Chicca A, Flores-Soto ME, Viveros-Paredes JM, Gertsch J. The Endocannabinoid Reuptake Inhibitor WOBE437 Is Orally Bioavailable and Exerts Indirect Polypharmacological Effects via Different Endocannabinoid Receptors. Front Mol Neurosci 2018; 11:180. [PMID: 29910713 PMCID: PMC5992379 DOI: 10.3389/fnmol.2018.00180] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022] Open
Abstract
Different anandamide (AEA) transport inhibitors show antinociceptive and antiinflammatory effects in vivo, but due to their concomitant inhibition of fatty acid amide hydrolase (FAAH) and overall poor bioavailability, they cannot be used unequivocally to study the particular role of endocannabinoid (EC) transport in pathophysiological conditions in vivo. Here, the potent and selective endocannabinoid reuptake inhibitor WOBE437, which inhibits AEA and 2-arachidonoylglycerol (2-AG) transport, was tested for its oral bioavailability to the brain. WOBE437 is assumed to locally increase EC levels in tissues in which facilitated EC reuptake intermediates subsequent hydrolysis. Given the marked polypharmacology of ECs, we hypothesized to see differential effects on distinct EC receptors in animal models of acute and chronic pain/inflammation. In C57BL6/J male mice, WOBE437 was orally bioavailable with an estimated tmax value of ≤20 min in plasma (Cmax ∼ 2000 pmol/mL after 50 mg/kg, p.o.) and brain (Cmax ∼ 500 pmol/g after 50 mg/kg, p.o.). WOBE437 was cleared from the brain after approximately 180 min. In addition, in BALB/c male mice, acute oral administration of WOBE437 (50 mg/kg) exhibited similar brain concentrations after 60 min and inhibited analgesia in the hot plate test in a cannabinoid CB1 receptor-dependent manner, without inducing catalepsy or affecting locomotion. WOBE437 significantly elevated AEA in the somatosensory cortex, while showing dose-dependent biphasic effects on 2-AG levels in plasma but no significant changes in N-acylethanolamines other than AEA in any of the tissues. In order to explore the presumed polypharmacology mediated via elevated EC levels, we tested this EC reuptake inhibitor in complete Freud's adjuvant induced monoarthritis in BALB/c mice as a model of chronic inflammation. Repetitive doses of WOBE437 (10 mg/kg, i.p.) attenuated allodynia and edema via cannabinoid CB2, CB1, and PPARγ receptors. The allodynia inhibition of WOBE437 treatment for 3 days was fully reversed by antagonists of any of the receptors. In the single dose treatment the CB2 and TRPV1 antagonists significantly blocked the effect of WOBE437. Overall, our results show the broad utility of WOBE437 for animal experimentation for both p.o. and i.p. administrations. Furthermore, the data indicate the possible involvement of EC reuptake/transport in pathophysiological processes related to pain and inflammation.
Collapse
Affiliation(s)
- Inés Reynoso-Moreno
- Institute of Biochemistry and Molecular Medicine, National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland.,Laboratorio de Investigación y Desarrollo Farmacéutico, Departamento de Farmacología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Mexico
| | - Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Mario E Flores-Soto
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Segura Social, Guadalajara, Mexico
| | - Juan M Viveros-Paredes
- Laboratorio de Investigación y Desarrollo Farmacéutico, Departamento de Farmacología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Mexico
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| |
Collapse
|
24
|
Bedse G, Bluett RJ, Patrick TA, Romness NK, Gaulden AD, Kingsley PJ, Plath N, Marnett LJ, Patel S. Therapeutic endocannabinoid augmentation for mood and anxiety disorders: comparative profiling of FAAH, MAGL and dual inhibitors. Transl Psychiatry 2018; 8:92. [PMID: 29695817 PMCID: PMC5917016 DOI: 10.1038/s41398-018-0141-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/28/2017] [Accepted: 02/22/2018] [Indexed: 12/23/2022] Open
Abstract
Recent studies have demonstrated anxiolytic potential of pharmacological endocannabinoid (eCB) augmentation approaches in a variety of preclinical models. Pharmacological inhibition of endocannabinoid-degrading enzymes, such as fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), elicit promising anxiolytic effects in rodent models with limited adverse behavioral effects, however, the efficacy of dual FAAH/MAGL inhibition has not been investigated. In the present study, we compared the effects of FAAH (PF-3845), MAGL (JZL184) and dual FAAH/MAGL (JZL195) inhibitors on (1) anxiety-like behaviors under non-stressed and stressed conditions, (2) locomotor activity and body temperature, (3) lipid levels in the brain and (4) cognitive functions. Behavioral analysis showed that PF-3845 or JZL184, but not JZL195, was able to prevent restraint stress-induced anxiety in the light-dark box assay when administered before stress exposure. Moreover, JZL195 treatment was not able to reverse foot shock-induced anxiety-like behavior in the elevated zero maze or light-dark box. JZL195, but not PF-3845 or JZL184, decreased body temperature and increased anxiety-like behavior in the open-field test. Overall, JZL195 did not show anxiolytic efficacy and the effects of JZL184 were more robust than that of PF-3845 in the models examined. These results showed that increasing either endogenous AEA or 2-AG separately produces anti-anxiety effects under stressful conditions but the same effects are not obtained from simultaneously increasing both AEA and 2-AG.
Collapse
Affiliation(s)
- Gaurav Bedse
- 0000 0004 1936 9916grid.412807.8Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN USA
| | - Rebecca J. Bluett
- 0000 0004 1936 9916grid.412807.8Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN USA ,0000 0001 2264 7217grid.152326.1Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN USA
| | - Toni A. Patrick
- 0000 0004 1936 9916grid.412807.8Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN USA
| | - Nicole K. Romness
- 0000 0004 1936 9916grid.412807.8Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN USA
| | - Andrew D. Gaulden
- 0000 0004 1936 9916grid.412807.8Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN USA
| | - Philip J. Kingsley
- 0000 0001 2264 7217grid.152326.1Departments 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 USA
| | - Niels Plath
- 0000 0004 0476 7612grid.424580.fH. Lundbeck A/S, Copenhagen, Denmark
| | - Lawrence J. Marnett
- 0000 0001 2264 7217grid.152326.1Departments 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 USA
| | - Sachin Patel
- 0000 0004 1936 9916grid.412807.8Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN USA ,0000 0001 2264 7217grid.152326.1Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN USA ,0000 0001 2264 7217grid.152326.1Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN USA
| |
Collapse
|
25
|
Sabban EL, Serova LI, Newman E, Aisenberg N, Akirav I. Changes in Gene Expression in the Locus Coeruleus-Amygdala Circuitry in Inhibitory Avoidance PTSD Model. Cell Mol Neurobiol 2018; 38:273-280. [PMID: 28889197 DOI: 10.1007/s10571-017-0548-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/02/2017] [Indexed: 12/28/2022]
Abstract
The locus coeruleus (LC)-amygdala circuit is implicated in playing a key role in responses to emotionally arousing stimuli and in the manifestation of post-traumatic stress disorder (PTSD). Here, we examined changes in gene expression of a number of important mediators of the LC-amygdala circuitry in the inhibition avoidance model of PTSD. After testing for basal acoustic startle response (ASR), rats were exposed to a severe footshock (1.5 mA for 10 s) in the inhibitory avoidance apparatus. They were given contextual situational reminders every 5 day for 25 days. Controls were treated identically but with the footshock inactivated. Animals were re-tested on second ASR and decapitated 1 h later. The shock group had enhanced hyperarousal and several changes in gene expression compared to controls. In the LC, mRNA levels of norepinephrine (NE) biosynthetic enzymes (TH, DBH), NE transporter (NET), NPY receptors (Y1R, Y2R), and CB1 receptor of endocannabinoid system were elevated. In the basolateral amygdala (BLA), there were marked reductions in gene expression for CB1, and especially Y1R, with rise for corticotropin-releasing hormone (CRH) system (CRH, CRH receptor 1), and no significant changes in the central amygdala. Our results suggest a fast forward mechanism in the LC-amygdala circuitry in the shock group.
Collapse
Affiliation(s)
- Esther L Sabban
- Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, Valhalla, NY, 10595, USA.
| | - Lidia I Serova
- Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, Valhalla, NY, 10595, USA
| | - Elizabeth Newman
- Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, Valhalla, NY, 10595, USA
| | - Nurit Aisenberg
- Department of Psychology, University of Haifa, 3498838, Haifa, Israel
| | - Irit Akirav
- Department of Psychology, University of Haifa, 3498838, Haifa, Israel
| |
Collapse
|
26
|
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: 154] [Impact Index Per Article: 25.7] [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
|
27
|
Balsevich G, Petrie GN, Hill MN. Endocannabinoids: Effectors of glucocorticoid signaling. Front Neuroendocrinol 2017; 47:86-108. [PMID: 28739508 DOI: 10.1016/j.yfrne.2017.07.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 01/17/2023]
Abstract
For decades, there has been speculation regarding the interaction of cannabinoids with glucocorticoid systems. Given the functional redundancy between many of the physiological effects of glucocorticoids and cannabinoids, it was originally speculated that the biological mechanisms of cannabinoids were mediated by direct interactions with glucocorticoid systems. With the discovery of the endocannabinoid system, additional research demonstrated that it was actually the opposite; glucocorticoids recruit endocannabinoid signaling, and that the engagement of endocannabinoid signaling mediated many of the neurobiological and physiological effects of glucocorticoids. With the development of advances in pharmacology and genetics, significant advances in this area have been made, and it is now clear that functional interactions between these systems are critical for a wide array of physiological processes. The current review acts a comprehensive summary of the contemporary state of knowledge regarding the biological interactions between glucocorticoids and endocannabinoids, and their potential role in health and disease.
Collapse
Affiliation(s)
- Georgia Balsevich
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Gavin N Petrie
- Hotchkiss Brain Institute, 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, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
28
|
Bedse G, Hartley ND, Neale E, Gaulden A, Patrick T, Kingsley P, Uddin MJ, Plath N, Marnett LJ, Patel S. Functional Redundancy Between Canonical Endocannabinoid Signaling Systems in the Modulation of Anxiety. Biol Psychiatry 2017; 82:488-499. [PMID: 28438413 PMCID: PMC5585044 DOI: 10.1016/j.biopsych.2017.03.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/13/2017] [Accepted: 03/06/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND Increasing the available repertoire of effective treatments for mood and anxiety disorders represents a critical unmet need. Pharmacological augmentation of endogenous cannabinoid (eCB) signaling has been suggested to represent a novel approach to the treatment of anxiety disorders; however, the functional interactions between two canonical eCB pathways mediated via anandamide (N-arachidonylethanolamine [AEA]) and 2-arachidonoylglycerol (2-AG) in the regulation of anxiety are not well understood. METHODS We utilized pharmacological augmentation and depletion combined with behavioral and electrophysiological approaches to probe the role of 2-AG signaling in the modulation of stress-induced anxiety and the functional redundancy between AEA and 2-AG signaling in the modulation of anxiety-like behaviors in mice. RESULTS Selective 2-AG augmentation reduced anxiety in the light/dark box assay and prevented stress-induced increases in anxiety associated with limbic AEA deficiency. In contrast, acute 2-AG depletion increased anxiety-like behaviors, which was normalized by selective pharmacological augmentation of AEA signaling and via direct cannabinoid receptor 1 stimulation with Δ9-tetrahydrocannabinol. Electrophysiological studies revealed 2-AG modulation of amygdala glutamatergic transmission as a key synaptic correlate of the anxiolytic effects of 2-AG augmentation. CONCLUSIONS Although AEA and 2-AG likely subserve distinct physiological roles, a pharmacological and functional redundancy between these canonical eCB signaling pathways exists in the modulation of anxiety-like behaviors. These data support development of eCB-based treatment approaches for mood and anxiety disorders and suggest a potentially wider therapeutic overlap between AEA and 2-AG augmentation approaches than was previously appreciated.
Collapse
Affiliation(s)
- Gaurav Bedse
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nolan D. Hartley
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Emily Neale
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew Gaulden
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Toni Patrick
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Philip 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, USA
| | - Md. Jashim Uddin
- 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, 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, USA
| | - Sachin Patel
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee.
| |
Collapse
|
29
|
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: 5.5] [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]
|
30
|
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: 410] [Impact Index Per Article: 51.3] [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
|
31
|
McLaughlin RJ, Verlezza S, Gray JM, Hill MN, Walker CD. Inhibition of anandamide hydrolysis dampens the neuroendocrine response to stress in neonatal rats subjected to suboptimal rearing conditions. Stress 2016; 19:114-24. [PMID: 26552023 DOI: 10.3109/10253890.2015.1117448] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Exposure to stress during early development can exert profound effects on the maturation of the neuroendocrine stress axis. The endocannabinoid (ECB) system has recently surfaced as a fundamental component of the neuroendocrine stress response; however, the effect of early-life stress on neonatal ECB signaling and the capacity to which ECB enhancement may modulate neonatal stress responses is relatively unknown. The present study assessed whether exposure to early-life stress in the form of limited access to nesting/bedding material (LB) from postnatal (PND) day 2 to 9 alters neuroendocrine activity and hypothalamic ECB content in neonatal rats challenged with a novel immobilization stressor. Furthermore, we examined whether inhibition of fatty acid amide hydrolase (FAAH), the enzyme responsible for the degradation of anandamide (AEA) affects neuroendocrine responses in PND10 pups as a function of rearing conditions. Neonatal rats showed a robust increase in corticosterone (CORT) and adrenocorticotropin hormone (ACTH) secretion in response to immobilization stress, which was significantly blunted in pups reared in LB conditions. Accordingly, LB pups exhibited reduced stress-induced Fos immunoreactivity in the paraventricular nucleus of the hypothalamus, with no significant differences in hypothalamic ECB content. Administration of the FAAH inhibitor URB597 (0.3 mg/kg, ip) 90 min prior to immobilization stress significantly dampened stress-induced CORT release, but only in pups reared in LB conditions. These results suggest that rearing in restricted bedding conditions dampens the neuroendocrine response to stress, while augmenting AEA mitigates stress-induced alterations in glucocorticoid secretion preferentially in pups subjected to early-life stress.
Collapse
Affiliation(s)
- Ryan Joseph McLaughlin
- a Douglas Mental Health University Institute, McGill University , Montreal , QC , Canada
- b Department of Integrative Physiology & Neuroscience , College of Veterinary Medicine, Washington State University , Pullman , WA , USA , and
| | - Silvanna Verlezza
- a Douglas Mental Health University Institute, McGill University , Montreal , QC , Canada
| | - Jennifer Megan Gray
- c Department of Cell Biology & Anatomy and Psychiatry , Hotchkiss Brain Institute, University of Calgary , Calgary , AB , Canada
| | - Matthew Nicholas Hill
- c Department of Cell Biology & Anatomy and Psychiatry , Hotchkiss Brain Institute, University of Calgary , Calgary , AB , Canada
| | | |
Collapse
|
32
|
Dickens MJ, Vecchiarelli HA, Hill MN, Bentley GE. Endocannabinoid Signaling in the Stress Response of Male and Female Songbirds. Endocrinology 2015; 156:4649-59. [PMID: 26431225 PMCID: PMC4655215 DOI: 10.1210/en.2015-1425] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Endocannabinoid (eCB) signaling plays an important role in the stress response pathways of the mammalian brain, yet its role in the avian stress response has not been described. Understanding eCB signaling in avian species (such as the European starling, Sturnus vulgaris) allows a model system that exhibits natural attenuation of hypothalamic-pituitary-adrenal (HPA) responsiveness to stressors. Specifically, seasonally breeding birds exhibit the highest HPA activity during the breeding season and subsequently exhibit a robust HPA down-regulation during molt. Because eCB signaling in mammals has an overall inhibitory effect on HPA activity, we expected shifts in eCB signaling to regulate the seasonal HPA down-regulation during molt. However, our data did not support a role for eCB signaling in the molt-related suppression of HPA activity. For example, injection of the cannabinoid receptor (CB1) antagonist, AM251, did not potentiate molt-suppressed HPA activity. Instead, our data suggest eCB regulation of HPA plasticity as birds transition from breeding to molt. In support of this hypothesis, birds in the late breeding season demonstrated a more dynamic response at the level of avian amygdala eCB content in response to acute stress. The response and directionality of this effect match that seen in mammals. Overall, our data suggest that eCB signaling may allow for a dynamic range in HPA responsiveness (eg, breeding), but the signaling pathway's role may be limited when the HPA response is restrained (eg, molt). This first characterization of eCB signaling in the avian stress response also emphasizes that although the system functions similarly to other species, its exact role may be species specific.
Collapse
Affiliation(s)
- Molly J Dickens
- Department of Integrative Biology (M.J.D., G.E.B.), University of California, Berkeley, Berkeley, California 94609; Department of Cell Biology and Anatomy (H.A.V., M.N.H.), Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4; and Helen Wills Neuroscience Institute (G.E.B.), University of California, Berkeley, Berkeley, California 94720-1650
| | - Haley A Vecchiarelli
- Department of Integrative Biology (M.J.D., G.E.B.), University of California, Berkeley, Berkeley, California 94609; Department of Cell Biology and Anatomy (H.A.V., M.N.H.), Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4; and Helen Wills Neuroscience Institute (G.E.B.), University of California, Berkeley, Berkeley, California 94720-1650
| | - Matthew N Hill
- Department of Integrative Biology (M.J.D., G.E.B.), University of California, Berkeley, Berkeley, California 94609; Department of Cell Biology and Anatomy (H.A.V., M.N.H.), Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4; and Helen Wills Neuroscience Institute (G.E.B.), University of California, Berkeley, Berkeley, California 94720-1650
| | - George E Bentley
- Department of Integrative Biology (M.J.D., G.E.B.), University of California, Berkeley, Berkeley, California 94609; Department of Cell Biology and Anatomy (H.A.V., M.N.H.), Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4; and Helen Wills Neuroscience Institute (G.E.B.), University of California, Berkeley, Berkeley, California 94720-1650
| |
Collapse
|
33
|
Tasker JG, Chen C, Fisher MO, Fu X, Rainville JR, Weiss GL. Endocannabinoid Regulation of Neuroendocrine Systems. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 125:163-201. [PMID: 26638767 DOI: 10.1016/bs.irn.2015.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The hypothalamus is a part of the brain that is critical for sustaining life through its homeostatic control and integrative regulation of the autonomic nervous system and neuroendocrine systems. Neuroendocrine function in mammals is mediated mainly through the control of pituitary hormone secretion by diverse neuroendocrine cell groups in the hypothalamus. Cannabinoid receptors are expressed throughout the hypothalamus, and endocannabinoids have been found to exert pronounced regulatory effects on neuroendocrine function via modulation of the outputs of several neuroendocrine systems. Here, we review the physiological regulation of neuroendocrine function by endocannabinoids, focusing on the role of endocannabinoids in the neuroendocrine regulation of the stress response, food intake, fluid homeostasis, and reproductive function. Cannabis sativa (marijuana) has a long history of recreational and/or medicinal use dating back to ancient times. It was used as an analgesic, anesthetic, and antianxiety herb as early as 2600 B.C. The hedonic, anxiolytic, and mood-elevating properties of cannabis have also been cited in ancient records from different cultures. However, it was not until 1964 that the psychoactive constituent of cannabis, Δ(9)-tetrahydrocannabinol, was isolated and its chemical structure determined (Gaoni & Mechoulam, 1964).
Collapse
Affiliation(s)
- Jeffrey G Tasker
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA; Neuroscience Program, Tulane University, New Orleans, Louisiana, USA.
| | - Chun Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA
| | - Marc O Fisher
- Neuroscience Program, Tulane University, New Orleans, Louisiana, USA
| | - Xin Fu
- Neuroscience Program, Tulane University, New Orleans, Louisiana, USA
| | - Jennifer R Rainville
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA
| | - Grant L Weiss
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
34
|
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: 2.9] [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
|
35
|
Abstract
PURPOSE OF REVIEW This review will provide updates on the neurobiology of stress, with specific focus on the transmitting mechanism of stress, the intricacies of integrating stress-related information, variability in response to stressors and clinical interventions. RECENT FINDINGS Neuropeptides have been implicated as a potential modulator for pathological stress via effects on neurogenesis, alterations in functional states of the cortico-limbic circuits and possible neuroprotection. Glucocorticoid receptors and mineralocorticoid receptors have been implicated in limbic modulation of stress and changes in the hypothalamic pituitary adrenal axis activity via corticosteroid signalling and feedback regulation. Chronic stress and genetic susceptibility have been found to promote intracellular mitochondrial and cellular DNA damage resulting in structural and functional alterations of neuroanatomy and neural circuitries. SUMMARY The interplay of stress and the neuroendocrine system appears to be influenced by alterations in genetic expression, resulting in neurochemical messenger and inflammatory cytokine alterations that impact neuroanatomy and neural circuitries to affect both central nervous system structure and function. These neurobiological contributions have been alluded to as possible contributors to stress-related psychopathologies. Clinical interventions with psychotropics, mind body therapies and behavioural therapies have been found to attenuate and reverse stress-related responses.
Collapse
Affiliation(s)
- Nadia Quijije
- Massachusetts General Hospital, Gregory Fricchione, MD Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
36
|
Evidence against a critical role of CB1 receptors in adaptation of the hypothalamic-pituitary-adrenal axis and other consequences of daily repeated stress. Eur Neuropsychopharmacol 2015; 25:1248-59. [PMID: 26092203 DOI: 10.1016/j.euroneuro.2015.04.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 03/24/2015] [Accepted: 04/26/2015] [Indexed: 02/01/2023]
Abstract
There is evidence that endogenous cannabinoids (eCBs) play a role in the control of the hypothalamic-pituitary-adrenal (HPA) axis, although they appear to have dual, stimulatory and inhibitory, effects. Recent data in rats suggest that eCBs, acting through CB1 receptors (CB1R), may be involved in adaptation of the HPA axis to daily repeated stress. In the present study we analyze this issue in male mice and rats. Using a knock-out mice for the CB1 receptor (CB1-/-) we showed that mutant mice presented similar adrenocorticotropic hormone (ACTH) response to the first IMO as wild-type mice. Daily repeated exposure to 1h of immobilization reduced the ACTH response to the stressor, regardless of the genotype, demonstrating that adaptation occurred to the same extent in absence of CB1R. Prototypical changes observed after repeated stress such as enhanced corticotropin releasing factor (CRH) gene expression in the paraventricular nucleus of the hypothalamus, impaired body weight gain and reduced thymus weight were similarly observed in both genotypes. The lack of effect of CB1R in the expression of HPA adaptation to another similar stressor (restraint) was confirmed in wild-type CD1 mice by the lack of effect of the CB1R antagonist AM251 just before the last exposure to stress. Finally, the latter drug did not blunt the HPA, glucose and behavioral adaptation to daily repeated forced swim in rats. Thus, the present results indicate that CB1R is not critical for overall effects of daily repeated stress or proper adaptation of the HPA axis in mice and rats.
Collapse
|
37
|
Lee TTY, Hill MN, Hillard CJ, Gorzalka BB. Disruption of peri-adolescent endocannabinoid signaling modulates adult neuroendocrine and behavioral responses to stress in male rats. Neuropharmacology 2015; 99:89-97. [PMID: 26192544 DOI: 10.1016/j.neuropharm.2015.07.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/14/2015] [Accepted: 07/16/2015] [Indexed: 10/23/2022]
Abstract
The endocannabinoid (eCB) system is known to regulate neural, endocrine and behavioral responses to stress in adults; however there is little knowledge regarding how this system governs the development and maturation of these responses. Previous work has reported dynamic and time-specific changes in CB1 receptor expression, N-arachidonylethanolamine (AEA) content and fatty acid amide hydrolase (FAAH) activity within corticolimbic structures throughout the peri-adolescent period. To examine whether fluctuations in adolescent eCB activity contribute to the development of adult stress responsivity and emotionality, we treated male Sprague-Dawley rats daily with the CB1R antagonist, AM-251 (5 mg/kg), or vehicle between post-natal days (PND) 35-45. Following this treatment, emotional behavior, HPA axis stress reactivity and habituation to repeated restraint stress, as well as corticolimbic eCB content were examined in adulthood (PND 75). Behaviorally, AM-251-treated males exhibited more active stress-coping behavior in the forced swim test, greater risk assessment behavior in the elevated plus maze and no significant differences in general motor activity. Peri-adolescent AM-251 treatment modified corticosterone habituation to repeated restraint exposure compared to vehicle. Peri-adolescent CB1R antagonism induced moderate changes in adult corticolimbic eCB signaling, with a significant decrease in amygdalar AEA, an increase in hypothalamic AEA and an increase in prefrontal cortical CB1R expression. Together, these data indicate that peri-adolescent endocannabinoid signaling contributes to the maturation of adult neurobehavioral responses to stress.
Collapse
Affiliation(s)
- Tiffany T-Y Lee
- Dept. of Psychology, University of British Columbia, Vancouver, V6T 1Z4, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, Dept. of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Cecilia J Hillard
- Dept. of Pharmacology and Toxicology and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Boris B Gorzalka
- Dept. of Psychology, University of British Columbia, Vancouver, V6T 1Z4, Canada.
| |
Collapse
|
38
|
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
|
39
|
Bedse G, Romano A, Tempesta B, Lavecchia MA, Pace L, Bellomo A, Duranti A, Micioni Di Bonaventura MV, Cifani C, Cassano T, Gaetani S. Inhibition of anandamide hydrolysis enhances noradrenergic and GABAergic transmission in the prefrontal cortex and basolateral amygdala of rats subjected to acute swim stress. J Neurosci Res 2015; 93:777-87. [DOI: 10.1002/jnr.23539] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 11/13/2014] [Accepted: 11/15/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Gaurav Bedse
- Department of Physiology and Pharmacology; Sapienza University of Rome; Rome Italy
| | - Adele Romano
- Department of Physiology and Pharmacology; Sapienza University of Rome; Rome Italy
| | - Bianca Tempesta
- Department of Physiology and Pharmacology; Sapienza University of Rome; Rome Italy
| | - Michele A. Lavecchia
- Department of Physiology and Pharmacology; Sapienza University of Rome; Rome Italy
| | - Lorenzo Pace
- Department of Clinical and Experimental Medicine; University of Foggia; Foggia Italy
| | | | - Andrea Duranti
- Department of Biomolecular Sciences; University of Urbino Carlo Bo; Urbino Italy
| | | | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino; Camerino Italy
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine; University of Foggia; Foggia Italy
| | - Silvana Gaetani
- Department of Physiology and Pharmacology; Sapienza University of Rome; Rome Italy
| |
Collapse
|
40
|
Ogawa S, Kunugi H. Inhibitors of Fatty Acid Amide Hydrolase and Monoacylglycerol Lipase: New Targets for Future Antidepressants. Curr Neuropharmacol 2015; 13:760-75. [PMID: 26630956 PMCID: PMC4759315 DOI: 10.2174/1570159x13666150612225212] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 03/16/2015] [Accepted: 03/18/2015] [Indexed: 12/27/2022] Open
Abstract
Cannabis and analogs of Δ<sup>9</sup>-tetrahydrocannabinol have been used for therapeutic purposes, but their therapeutic use remains limited because of various adverse effects. Endogenous cannabinoids have been discovered, and dysregulation of endocannabinoid signaling is implicated in the pathophysiology of major depressive disorder (MDD). Recently, endocannabinoid hydrolytic enzymes such as fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) have become new therapeutic targets in the treatment of MDD. Several FAAH or MAGL inhibitors are reported to have no cannabimimetic side effects and, therefore, are new potential therapeutic options for patients with MDD who are resistant to first-line antidepressants (selective serotonin and serotonin-norepinephrine reuptake inhibitors). In this review, we focus on the possible relationships between MDD and the endocannabinoid system as well as the inhibitors' therapeutic potential. MAGL inhibitors may reduce inflammatory responses through activation of cannabinoid receptor type 2. In the hypothalamic-pituitary-adrenal axis, repeated FAAH inhibitor administration may be beneficial for reducing circulating glucocorticoid levels. Both FAAH and MAGL inhibitors may contribute to dopaminergic system regulation. Recently, several new inhibitors have been developed with strong potency and selectivity. FAAH inhibitor, MAGL inhibitor, or dual blocker use would be promising new treatments for MDD. Further pre-clinical studies and clinical trials using these inhibitors are warranted.
Collapse
Affiliation(s)
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| |
Collapse
|