1
|
Borland JM. A review of the effects of different types of social behaviors on the recruitment of neuropeptides and neurotransmitters in the nucleus accumbens. Front Neuroendocrinol 2025; 77:101175. [PMID: 39892577 DOI: 10.1016/j.yfrne.2025.101175] [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: 05/04/2024] [Revised: 01/25/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
There is a lack of understanding of the neural mechanisms regulating the rewarding effects of social interactions. A significant contributor to this lack of clarity is the diversity of social behaviors and animal models utilized to investigate mechanisms. Other sources of the lack of clarity are the diversity of brain regions that can regulate social reward and the diversity of signaling pathways that regulate reward. To provide some clarity into the mechanisms of social reward, this review focused on the brain region most implicated in reward for multiple stimuli, the nucleus accumbens, and surveyed (systematically reviewed) studies that investigated the relationship between social interaction and five signaling systems implicated in the regulation of reward and social behavior: oxytocin, vasopressin, serotonin, opioids and endocannabinoids. Moreover, all of these studies were organized by the type of social behavior studied: affiliative interactions, play behavior, aggression, social defeat, sex behavior, pair-bonding, parental behavior and social isolation. From this survey and organization, this review concludes that oxytocin, endocannabinoids and mu-opioid receptors in the nucleus accumbens positively regulate the rewarding social behaviors, and kappa-opioid receptors negatively regulate the rewarding social behaviors. The opposite profile is observed for these signaling systems for the aversive social behaviors. More studies are needed to investigate the directional role of the serotonin system in the nucleus accumbens in the regulation of many types of social behaviors, and vasopressin likely does not act in the nucleus accumbens in the regulation of the valence of social behaviors. Many of these different signaling systems are also interdependent of one another in the regulation of different types of social behaviors. Finally, the interaction of these signaling systems with dopamine in the nucleus accumbens is briefly discussed.
Collapse
|
2
|
Jana A, Nath A, Sen P, Kundu S, Alghamdi BS, Abujamel TS, Saboor M, Woon-Khiong C, Alexiou A, Papadakis M, Alam MZ, Ashraf GM. Unraveling the Endocannabinoid System: Exploring Its Therapeutic Potential in Autism Spectrum Disorder. Neuromolecular Med 2024; 26:20. [PMID: 38744725 PMCID: PMC11093854 DOI: 10.1007/s12017-024-08781-6] [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: 12/08/2023] [Accepted: 03/04/2024] [Indexed: 05/16/2024]
Abstract
The salient features of autism spectrum disorder (ASD) encompass persistent difficulties in social communication, as well as the presence of restricted and repetitive facets of behavior, hobbies, or pursuits, which are often accompanied with cognitive limitations. Over the past few decades, a sizable number of studies have been conducted to enhance our understanding of the pathophysiology of ASD. Preclinical rat models have proven to be extremely valuable in simulating and analyzing the roles of a wide range of established environmental and genetic factors. Recent research has also demonstrated the significant involvement of the endocannabinoid system (ECS) in the pathogenesis of several neuropsychiatric diseases, including ASD. In fact, the ECS has the potential to regulate a multitude of metabolic and cellular pathways associated with autism, including the immune system. Moreover, the ECS has emerged as a promising target for intervention with high predictive validity. Particularly noteworthy are resent preclinical studies in rodents, which describe the onset of ASD-like symptoms after various genetic or pharmacological interventions targeting the ECS, providing encouraging evidence for further exploration in this area.
Collapse
Affiliation(s)
- Ankit Jana
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Arnab Nath
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Palash Sen
- School of Biosciences, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Swikriti Kundu
- Siksha Bhavana, Visva-Bharati University, Bolpur, West Bengal, 731235, India
| | - Badrah S Alghamdi
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Turki S Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad Saboor
- Department of Medical Laboratory Sciences, College of Health Sciences, and Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Chan Woon-Khiong
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, Greece
- Department of Research & Development, AFNP Med, 1030, Vienna, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohammad Zubair Alam
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, and Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
3
|
Therapeutic Molecular Insights into the Active Engagement of Cannabinoids in the Therapy of Parkinson's Disease: A Novel and Futuristic Approach. Neurotox Res 2023; 41:85-102. [PMID: 36567416 DOI: 10.1007/s12640-022-00619-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/09/2022] [Accepted: 12/05/2022] [Indexed: 12/27/2022]
Abstract
Parkinson's disease is a neurodegenerative disorder which is characterised mostly by loss of dopaminergic nerve cells throughout the nigral area mainly as a consequence of oxidative stress. Muscle stiffness, disorganised bodily responses, disturbed sleep, weariness, amnesia, and voice impairment are all symptoms of dopaminergic neuron degeneration and existing symptomatic treatments are important to arrest additional neuronal death. Some cannabinoids have recently been demonstrated as robust antioxidants that might protect the nerve cells from degeneration even when cannabinoid receptors are not triggered. Cannabinoids are likely to have property to slow or presumably cease the steady deterioration of the brain's dopaminergic systems, a condition for which there is now no treatment. The use of cannabinoids in combination with currently available drugs has the potential to introduce a radically new paradigm for treatment of Parkinson's disease, making it immensely useful in the treatment of such a debilitating illness.
Collapse
|
4
|
Calakos KC, Liu H, Lu Y, Anderson JM, Matuskey D, Nabulsi N, Ye Y, Skosnik PD, D'Souza DC, Morris ED, Cosgrove KP, Hillmer AT. Assessment of transient dopamine responses to smoked cannabis. Drug Alcohol Depend 2021; 227:108920. [PMID: 34399137 PMCID: PMC8464527 DOI: 10.1016/j.drugalcdep.2021.108920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/14/2021] [Accepted: 06/08/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Dopaminergic mechanisms that may underlie cannabis' reinforcing effects are not well elucidated in humans. This positron emission tomography (PET) imaging study used the dopamine D2/3 receptor antagonist [11C]raclopride and kinetic modelling testing for transient changes in radiotracer uptake to assess the striatal dopamine response to smoked cannabis in a preliminary sample. METHODS PET emission data were acquired from regular cannabis users (n = 14; 7 M/7 F; 19-32 years old) over 90 min immediately after [11C]raclopride administration (584 ± 95 MBq) as bolus followed by constant infusion (Kbol = 105 min). Participants smoked a cannabis cigarette, using a paced puff protocol, 35 min after scan start. Plasma concentrations of Δ9-THC and metabolites and ratings of subjective "high" were collected during imaging. Striatal dopamine responses were assessed voxelwise with a kinetic model testing for transient reductions in [11C]raclopride binding, linear-parametric neurotransmitter PET (lp-ntPET) (cerebellum as a reference region). RESULTS Cannabis smoking increased plasma Δ9-THC levels (peak: 0-10 min) and subjective high (peak: 0-30 min). Significant clusters (>16 voxels) modeled by transient reductions in [11C]raclopride binding were identified for all 12 analyzed scans. In total, 26 clusters of significant responses to cannabis were detected, of which 16 were located in the ventral striatum, including at least one ventral striatum cluster in 11 of the 12 analyzed scans. CONCLUSIONS These preliminary data support the sensitivity of [11C]raclopride PET with analysis of transient changes in radiotracer uptake to detect cannabis smoking-induced dopamine responses. This approach shows future promise to further elucidate roles of mesolimbic dopaminergic signaling in chronic cannabis use. ClinicalTrials.gov Identifier: NCT02817698.
Collapse
Affiliation(s)
- Katina C Calakos
- Department of Psychiatry, Yale School of Medicine, 300 George Street, New Haven, CT, 06511, United States; Interdepartmental Neuroscience Program, Yale University, 333 Cedar Street, New Haven, CT, 06510, United States.
| | - Heather Liu
- Department of Biomedical Engineering, Yale University, 17 Hillhouse Avenue, New Haven, CT, 06511, United States.
| | - Yihuan Lu
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 330 Cedar Street, New Haven, CT, 06519, United States; Yale PET Center, Yale University, 801 Howard Avenue, New Haven, CT, 06510, United States.
| | - Jon Mikael Anderson
- Department of Psychiatry, Yale School of Medicine, 300 George Street, New Haven, CT, 06511, United States.
| | - David Matuskey
- Department of Psychiatry, Yale School of Medicine, 300 George Street, New Haven, CT, 06511, United States; Department of Radiology and Biomedical Imaging, Yale School of Medicine, 330 Cedar Street, New Haven, CT, 06519, United States; Yale PET Center, Yale University, 801 Howard Avenue, New Haven, CT, 06510, United States; Department of Neurology, Yale School of Medicine, 800 Howard Avenue, New Haven, CT, 06519, United States.
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 330 Cedar Street, New Haven, CT, 06519, United States; Yale PET Center, Yale University, 801 Howard Avenue, New Haven, CT, 06510, United States.
| | - Yunpeng Ye
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 330 Cedar Street, New Haven, CT, 06519, United States; Yale PET Center, Yale University, 801 Howard Avenue, New Haven, CT, 06510, United States.
| | - Patrick D Skosnik
- Department of Psychiatry, Yale School of Medicine, 300 George Street, New Haven, CT, 06511, United States; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, 34 Park Street, New Haven, CT, 06519, United States.
| | - Deepak Cyril D'Souza
- Department of Psychiatry, Yale School of Medicine, 300 George Street, New Haven, CT, 06511, United States; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, 34 Park Street, New Haven, CT, 06519, United States.
| | - Evan D Morris
- Department of Psychiatry, Yale School of Medicine, 300 George Street, New Haven, CT, 06511, United States; Interdepartmental Neuroscience Program, Yale University, 333 Cedar Street, New Haven, CT, 06510, United States; Department of Biomedical Engineering, Yale University, 17 Hillhouse Avenue, New Haven, CT, 06511, United States; Department of Radiology and Biomedical Imaging, Yale School of Medicine, 330 Cedar Street, New Haven, CT, 06519, United States; Yale PET Center, Yale University, 801 Howard Avenue, New Haven, CT, 06510, United States.
| | - Kelly P Cosgrove
- Department of Psychiatry, Yale School of Medicine, 300 George Street, New Haven, CT, 06511, United States; Interdepartmental Neuroscience Program, Yale University, 333 Cedar Street, New Haven, CT, 06510, United States; Department of Radiology and Biomedical Imaging, Yale School of Medicine, 330 Cedar Street, New Haven, CT, 06519, United States; Department of Neuroscience, Yale University, 333 Cedar Street, New Haven, CT, 06510, United States.
| | - Ansel T Hillmer
- Department of Psychiatry, Yale School of Medicine, 300 George Street, New Haven, CT, 06511, United States; Department of Biomedical Engineering, Yale University, 17 Hillhouse Avenue, New Haven, CT, 06511, United States; Department of Radiology and Biomedical Imaging, Yale School of Medicine, 330 Cedar Street, New Haven, CT, 06519, United States; Yale PET Center, Yale University, 801 Howard Avenue, New Haven, CT, 06510, United States.
| |
Collapse
|
5
|
de Sa Nogueira D, Bourdy R, Filliol D, Awad G, Andry V, Goumon Y, Olmstead MC, Befort K. Binge sucrose-induced neuroadaptations: A focus on the endocannabinoid system. Appetite 2021; 164:105258. [PMID: 33864862 DOI: 10.1016/j.appet.2021.105258] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/24/2021] [Accepted: 04/06/2021] [Indexed: 12/26/2022]
Abstract
Binge eating, the defining feature of binge eating disorder (BED), is associated with a number of adverse health outcomes as well as a reduced quality of life. Animals, like humans, selectively binge on highly palatable food suggesting that the behaviour is driven by hedonic, rather than metabolic, signals. Given the links to both reward processing and food intake, this study examined the contribution of the endocannabinoid system (ECS) to binge-like eating in rats. Separate groups were given intermittent (12 h) or continuous (24 h) access to 10% sucrose and food over 28 days, with only the 12 h access group displaying excessive sucrose intake within a discrete period of time (i.e., binge eating). Importantly, this group also exhibited alterations in ECS transcripts and endocannabinoid levels in brain reward regions, including an increase in cannabinoid receptor 1 (CB1R) mRNA in the nucleus accumbens as well as changes in endocannabinoid levels in the prefrontal cortex and hippocampus. We then tested whether different doses (1 and 3 mg/kg) of a CB1R antagonist, Rimonabant, modify binge-like intake or the development of a conditioned place preference (CPP) to sucrose. CB1R blockade reduced binge-like intake of sucrose and blocked a sucrose CPP, but only in rats that had undergone 28 days of sucrose consumption. These findings indicate that sucrose bingeing alters the ECS in reward-related areas, modifications that exacerbate the effect of CB1R blockade on sucrose reward. Overall, our results broaden the understanding of neural alterations associated with bingeing eating and demonstrate an important role for CB1R mechanisms in reward processing. In addition, these findings have implications for understanding substance abuse, which is also characterized by excessive and maladaptive intake, pointing towards addictive-like properties of palatable food.
Collapse
Affiliation(s)
- David de Sa Nogueira
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, 12 rue Goethe, F-67000, Strasbourg France; Current Address: Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Romain Bourdy
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, 12 rue Goethe, F-67000, Strasbourg France
| | - Dominique Filliol
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, 12 rue Goethe, F-67000, Strasbourg France
| | - Gaëlle Awad
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, 12 rue Goethe, F-67000, Strasbourg France
| | - Virginie Andry
- Institut des Neurosciences Cellulaires et Intégratives (INCI), UPR 3212, CNRS, 8 Allée du Général Rouvillois, 67000, Strasbourg, France
| | - Yannick Goumon
- Institut des Neurosciences Cellulaires et Intégratives (INCI), UPR 3212, CNRS, 8 Allée du Général Rouvillois, 67000, Strasbourg, France
| | - Mary C Olmstead
- Department of Psychology, Center for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Katia Befort
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, 12 rue Goethe, F-67000, Strasbourg France.
| |
Collapse
|
6
|
Crane NA, Phan KL. Effect of Δ9-Tetrahydrocannabinol on frontostriatal resting state functional connectivity and subjective euphoric response in healthy young adults. Drug Alcohol Depend 2021; 221:108565. [PMID: 33592558 PMCID: PMC8026570 DOI: 10.1016/j.drugalcdep.2021.108565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/16/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Few studies have examined how Δ9-Tetrahydrocannabinol (THC), the main psychoactive component of cannabis, impacts brain reward circuitry in humans. In this study, we examined if an acute dose of THC altered resting state functional connectivity between the striatum and prefrontal cortex among healthy young adults with limited cannabis use. METHODS Participants received THC (n = 24) or placebo (n = 22) in a double-blind, randomized, between-subject design. Participants completed self-report measures of euphoria and drug-liking throughout the visit. Approximately 120 min after drug administration, participants completed an 8-min resting state functional MRI (rs-fMRI) scan. We utilized seed-based connectivity of the striatum (bilateral putamen, caudate, and NAcc seeds) to the frontal cortex. RESULTS Individuals who received THC demonstrated greater rs-fMRI connectivity between the right NAcc and regions of the medial prefrontal cortex (mPFC) (p-values<0.05, corrected) and higher subjective euphoria ratings (p = .03) compared to compared to individuals who received placebo. Higher ratings of euphoria were related to greater right NAcc-dorsal mPFC (dmPFC) connectivity for the THC group (p=.03), but not for the placebo group (p=.98). CONCLUSIONS This is one of the first studies to examine rs-fMRI connectivity in healthy young non-users after THC administration. We found individuals receiving THC show greater rs-fMRI connectivity between the NAcc and mPFC, regions implicated in reward, compared to individuals receiving placebo. In addition, individuals receiving THC reported higher subjective euphoria ratings, which were positively associated with NAcc-dmPFC connectivity. Overall, our findings suggest THC may produce subjective and neural reward responses that contribute to the rewarding, reinforcing properties of cannabis.
Collapse
Affiliation(s)
| | - K. Luan Phan
- Department of Psychiatry, University of Illinois at
Chicago,Department of Psychiatry and Behavioral Health, The Ohio
State University
| |
Collapse
|
7
|
Su T, Yan Y, Li Q, Ye J, Pei L. Endocannabinoid System Unlocks the Puzzle of Autism Treatment via Microglia. Front Psychiatry 2021; 12:734837. [PMID: 34744824 PMCID: PMC8568770 DOI: 10.3389/fpsyt.2021.734837] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/27/2021] [Indexed: 01/15/2023] Open
Abstract
Autism spectrum disorder (ASD) is a serious neurodevelopmental disorder and characterized by early childhood-onset impairments in social interaction and communication, restricted and repetitive patterns of behavior or interests. So far there is no effective treatment for ASD, and the pathogenesis of ASD remains unclear. Genetic and epigenetic factors have been considered to be the main cause of ASD. It is known that endocannabinoid and its receptors are widely distributed in the central nervous system, and provide a positive and irreversible change toward a more physiological neurodevelopment. Recently, the endocannabinoid system (ECS) has been found to participate in the regulation of social reward behavior, which has attracted considerable attention from neuroscientists and neurologists. Both animal models and clinical studies have shown that the ECS is a potential target for the treatment of autism, but the mechanism is still unknown. In the brain, microglia express a complete ECS signaling system. Studies also have shown that modulating ECS signaling can regulate the functions of microglia. By comprehensively reviewing previous studies and combining with our recent work, this review addresses the effects of targeting ECS on microglia, and how this can contribute to maintain the positivity of the central nervous system, and thus improve the symptoms of autism. This will provide insights for revealing the mechanism and developing new treatment strategies for autism.
Collapse
Affiliation(s)
- Tangfeng Su
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Yan
- Department of Neurology, People's Hospital of Dongxihu District, Wuhan, China
| | - Qiang Li
- Exchange, Development and Service Center for Science and Technology Talents, The Ministry of Science and Technology, Beijing, China
| | - Jiacai Ye
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Lei Pei
- Collaborative Innovation Center for Brain Science, The Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, China.,Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
8
|
Cannabis Extract Composition Determines Reinforcement in a Vapor Self-Administration Paradigm. J Neurosci 2020; 40:6264-6266. [PMID: 32801126 DOI: 10.1523/jneurosci.0814-20.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/21/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023] Open
|
9
|
Vaseghi S, Nasehi M, Zarrindast MR. How do stupendous cannabinoids modulate memory processing via affecting neurotransmitter systems? Neurosci Biobehav Rev 2020; 120:173-221. [PMID: 33171142 DOI: 10.1016/j.neubiorev.2020.10.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/17/2020] [Accepted: 10/26/2020] [Indexed: 12/27/2022]
Abstract
In the present study, we wanted to review the role of cannabinoids in learning and memory in animal models, with respect to their interaction effects with six principal neurotransmitters involved in learning and memory including dopamine, glutamate, GABA (γ-aminobutyric acid), serotonin, acetylcholine, and noradrenaline. Cannabinoids induce a wide-range of unpredictable effects on cognitive functions, while their mechanisms are not fully understood. Cannabinoids in different brain regions and in interaction with different neurotransmitters, show diverse responses. Previous findings have shown that cannabinoids agonists and antagonists induce various unpredictable effects such as similar effect, paradoxical effect, or dualistic effect. It should not be forgotten that brain neurotransmitter systems can also play unpredictable roles in mediating cognitive functions. Thus, we aimed to review and discuss the effect of cannabinoids in interaction with neurotransmitters on learning and memory. In addition, we mentioned to the type of interactions between cannabinoids and neurotransmitter systems. We suggested that investigating the type of interactions is a critical neuropharmacological issue that should be considered in future studies.
Collapse
Affiliation(s)
- Salar Vaseghi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohammad-Reza Zarrindast
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Meneses-Gaya CD, Crippa JA, Hallak JE, Miguel AQ, Laranjeira R, Bressan RA, Zuardi AW, Lacerda AL. Cannabidiol for the treatment of crack-cocaine craving: an exploratory double-blind study. BRAZILIAN JOURNAL OF PSYCHIATRY 2020; 43:467-476. [PMID: 33146345 PMCID: PMC8555645 DOI: 10.1590/1516-4446-2020-1416] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022]
Abstract
Objective: To assess the efficacy of cannabidiol (CBD) in the management of crack-cocaine craving and the treatment of frequent withdrawal symptoms. Methods: Thirty-one men with a diagnosis of crack-cocaine dependence were enrolled in a randomized, double-blind, placebo-controlled trial. We applied neuropsychological tests and assessed craving intensity, anxiety and depression symptoms, and substance use patterns at baseline and at the end of the trial. The participants were treated with CBD 300 mg/day or placebo for 10 days. During this period, we used a technique to induce craving and assessed the intensity of symptoms before and after the induction procedure. Results: Craving levels reduced significantly over the 10 days of the trial, although no differences were found between the CBD and placebo groups. Craving induction was successful in both groups, with no significant differences between them. Indicators of anxiety, depression, and sleep alterations before and after treatment also did not differ across groups. Conclusion: Under the conditions of this trial, CBD was unable to interfere with symptoms of crack-cocaine withdrawal. Further studies with larger outpatient samples involving different doses and treatment periods would be desirable and timely to elucidate the potential of CBD to induce reductions in crack-cocaine self-administration.
Collapse
Affiliation(s)
- Carolina de Meneses-Gaya
- Laboratório Interdisciplinar de Neurociências Clínicas (LiNC), Departamento de Psiquiatria, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - José A. Crippa
- Divisão de Psiquiatria, Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Jaime E. Hallak
- Divisão de Psiquiatria, Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - André Q. Miguel
- Instituto Nacional de Políticas Públicas do Álcool e Drogas (INPAD), UNIFESP, Brazil
| | - Ronaldo Laranjeira
- Instituto Nacional de Políticas Públicas do Álcool e Drogas (INPAD), UNIFESP, Brazil
| | - Rodrigo A. Bressan
- Laboratório Interdisciplinar de Neurociências Clínicas (LiNC), Departamento de Psiquiatria, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - Antonio W. Zuardi
- Divisão de Psiquiatria, Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Acioly L. Lacerda
- Laboratório Interdisciplinar de Neurociências Clínicas (LiNC), Departamento de Psiquiatria, Universidade Federal de São Paulo (UNIFESP), Brazil
| |
Collapse
|
11
|
Abstract
Addiction is commonly identified with habitual nonmedical self-administration of drugs. It is usually defined by characteristics of intoxication or by characteristics of withdrawal symptoms. Such addictions can also be defined in terms of the brain mechanisms they activate; most addictive drugs cause elevations in extracellular levels of the neurotransmitter dopamine. Animals unable to synthesize or use dopamine lack the conditioned reflexes discussed by Pavlov or the appetitive behavior discussed by Craig; they have only unconditioned consummatory reflexes. Burst discharges (phasic firing) of dopamine-containing neurons are necessary to establish long-term memories associating predictive stimuli with rewards and punishers. Independent discharges of dopamine neurons (tonic or pacemaker firing) determine the motivation to respond to such cues. As a result of habitual intake of addictive drugs, dopamine receptors expressed in the brain are decreased, thereby reducing interest in activities not already stamped in by habitual rewards.
Collapse
Affiliation(s)
- Roy A Wise
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, USA; .,Behavioral Genetics Laboratory, McLean Hospital, Belmont, Massachusetts 02478, USA;
| | - Mykel A Robble
- Behavioral Genetics Laboratory, McLean Hospital, Belmont, Massachusetts 02478, USA;
| |
Collapse
|
12
|
Hauser SR, Katner SN, Waeiss RA, Truitt WA, Bell RL, McBride WJ, Rodd ZA. Selective breeding for high alcohol preference is associated with increased sensitivity to cannabinoid reward within the nucleus accumbens shell. Pharmacol Biochem Behav 2020; 197:173002. [PMID: 32710885 DOI: 10.1016/j.pbb.2020.173002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 11/30/2022]
Abstract
RATIONALE The rate of cannabinoid intake by those with alcohol use disorder (AUD) exceeds that of the general public. The high prevalence of co-abuse of alcohol and cannabis has been postulated to be predicated upon both a common predisposing genetic factor and the interaction of the drugs within the organism. The current experiments examined the effects of cannabinoids in an animal model of AUD. OBJECTIVES The present study assessed the reinforcing properties of a cannabinoid receptor 1 (CB1) agonist self-administered directly into the nucleus accumbens shell (AcbSh) in female Wistar and alcohol-preferring (P) rats. METHODS Following guide cannulae surgery aimed at AcbSh, subjects were placed in an operant box equipped with an 'active lever' (fixed ratio 1; FR1) that caused the delivery of the infusate and an 'inactive lever' that did not. Subjects were arbitrarily assigned to one of seven groups that self-administered either artificial cerebrospinal fluid (aCSF), or 3.125, 6.25, 12.5, or 25 pmol/100 nl of O-1057, a water-soluble CB1 agonist, dissolved in aCSF. The first four sessions of acquisition are followed by aCSF only infusates in sessions 5 and 6 during extinction, and finally the acquisition dose of infusate during session 7 as reinstatement. RESULTS The CB1 agonist was self-administered directly into the AcbSh. P rats self-administered the CB1 agonist at lower concentrations and at higher rates compared to Wistar rats. CONCLUSIONS Overall, the data indicate selective breeding for high alcohol preference has produced rats divergent in response to cannabinoids within the brain reward pathway. The data support the hypothesis that there can be common genetic factors influencing drug addiction.
Collapse
Affiliation(s)
- Sheketha R Hauser
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Simon N Katner
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Robert A Waeiss
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - William A Truitt
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Richard L Bell
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - William J McBride
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zachary A Rodd
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
13
|
Feja M, Leigh MPK, Baindur AN, McGraw JJ, Wakabayashi KT, Cravatt BF, Bass CE. The novel MAGL inhibitor MJN110 enhances responding to reward-predictive incentive cues by activation of CB1 receptors. Neuropharmacology 2020; 162:107814. [PMID: 31628934 PMCID: PMC6983961 DOI: 10.1016/j.neuropharm.2019.107814] [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: 07/27/2018] [Revised: 10/14/2019] [Accepted: 10/14/2019] [Indexed: 11/17/2022]
Abstract
CB1 receptor antagonists disrupt operant responding for food and drug reinforcers, and cue-induced reinstatement of cocaine and heroin seeking. Conversely, enhancing endocannabinoid signaling, particularly 2-arachidonyl glycerol (2-AG), by inhibition of monoacyl glycerol lipase (MAGL), may facilitate some aspects of reward seeking. To determine how endocannabinoid signaling affects responding to reward-predictive cues, we employed an operant task that allows us to parse the incentive motivational properties of cues. Rats were required to nosepoke during an intermittent audiovisual incentive cue (IC) to obtain a 10% sucrose reward. The CB1 receptor antagonist, rimonabant, dose-dependently decreased the response ratio (rewarded ICs/total presented) and active nosepokes per IC, while it increased the latency to respond to the cue and obtain the reward, indicating an overall decrease in both the choice and vigor of responding. Yet rats persisted in entering the reward cup. Using a modified version of the task, the novel MAGL inhibitor MJN110 increased the response ratio, decreased the latencies to respond to the IC and enhanced active nosepokes per IC, indicating a facilitation of cue-induced reward seeking. These effects were blocked by a subthreshold dose of rimonabant. Finally, MJN110 did not alter consumption of freely available sucrose within volumes obtained in the operant task. Together these data demonstrate blocking endocannabinoid tone at the CB1 receptor attenuates the ability of cues to induce reward seeking, while some aspects of motivation for the reward are retained. Conversely, enhancing 2-AG signaling at CB1 receptors facilitates IC responding and increases the motivational properties of the IC.
Collapse
Affiliation(s)
- Malte Feja
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main St, Buffalo, NY, 14203, USA.
| | - Martin P K Leigh
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main St, Buffalo, NY, 14203, USA.
| | - Ajay N Baindur
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main St, Buffalo, NY, 14203, USA.
| | - Justin J McGraw
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main St, Buffalo, NY, 14203, USA.
| | - Ken T Wakabayashi
- Department of Psychology, University of Nebraska-Lincoln, 1220 T. Street, Lincoln, NE, 68503, USA.
| | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - Caroline E Bass
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main St, Buffalo, NY, 14203, USA.
| |
Collapse
|
14
|
Bán EG, Brassai A, Vizi ES. The role of the endogenous neurotransmitters associated with neuropathic pain and in the opioid crisis: The innate pain-relieving system. Brain Res Bull 2019; 155:129-136. [PMID: 31816407 DOI: 10.1016/j.brainresbull.2019.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/22/2019] [Accepted: 12/02/2019] [Indexed: 12/28/2022]
Abstract
Neuropathic pain is a chronic pain caused by central and peripheral nerve injury, long-term diabetes or treatment with chemotherapy drugs, and it is dissimilar to other chronic pain conditions. Chronic pain usually seriously affects the quality of life, and its drug treatment may result in increased costs of social and medical care. As in the USA and Canada, in Europe, the demand for pain-relieving medicines used in chronic pain has also significantly increased, but most European countries are not experiencing an opioid crisis. In this review, the role of various endogenous transmitters (noradrenaline, dopamine, serotonin, met- and leu-enkephalins, β-endorphin, dynorphins, cannabinoids, ATP) and various receptors (α2, μ, etc.) in the innate pain-relieving system will be discussed. Furthermore, the modulation of pain processing pathways by transmitters, focusing on neuropathic pain and the role of the sympathetic nervous system in the side effects of excessive opioid treatment, will be explained.
Collapse
Affiliation(s)
- E Gy Bán
- Dept. ME1, Faculty of Medicine in English, "George Emil Palade" University of Medicine, Pharmacy, Science and Technology of Târgu-Mureș, Marosvásárhely, Romania
| | - A Brassai
- Dept. ME1, Faculty of Medicine in English, "George Emil Palade" University of Medicine, Pharmacy, Science and Technology of Târgu-Mureș, Marosvásárhely, Romania
| | - E S Vizi
- Institute of Experimental Medicine, Budapest, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
15
|
Baul HS, Manikandan C, Sen D. Cannabinoid receptor as a potential therapeutic target for Parkinson's Disease. Brain Res Bull 2019; 146:244-252. [PMID: 30664919 DOI: 10.1016/j.brainresbull.2019.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 01/07/2019] [Accepted: 01/14/2019] [Indexed: 11/16/2022]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, characterized by the loss of dopaminergic neurons from substantia nigra pars compacta of basal ganglia caused due to gene mutation, misfolded protein aggregation, reactive oxygen species generation and inflammatory stress. Degeneration of dopaminergic neurons results in muscle stiffness, uncoordinated body movements, sleep disturbance, fatigue, amnesia and impaired voice. Currently, levodopa (L-DOPA) administration is the most widely used therapy for PD. But prolonged administration of L-DOPA is associated with the symptoms of dyskinesia. However, emerging evidences suggest the role of cannabinoid receptors (CBRs) in curtailing the progression of PD by activating neuroprotective pathways. Hence, cannabinoid therapy could be a promising alternative to combat PD in future. In the present review we have discussed the potential role of CBRs in attenuating the key mechanisms of PD and how the existing research gaps needs to be bridged in order to understand the molecular mechanism of CBRs in detail.
Collapse
Affiliation(s)
- Himadri Shekhaar Baul
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - Ceera Manikandan
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - Dwaipayan Sen
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
16
|
Acupuncture on the Stress-Related Drug Relapse to Seeking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:5367864. [PMID: 30416533 PMCID: PMC6207895 DOI: 10.1155/2018/5367864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 01/30/2018] [Accepted: 07/19/2018] [Indexed: 11/23/2022]
Abstract
Drug addiction is a chronic relapsing disease, which causes serious social and economic problems. The most important trial for the successful treatment of drug addiction is to prevent the high rate of relapse to drug-seeking behaviors. Opponent process as a motivational theory with excessive drug seeking in the negative reinforcement of drug dependence reflects both loss of brain reward system and recruitment of brain stress system. The negative emotional state produced by brain stress system during drug withdrawal might contribute to the intense drug craving and drive drug-seeking behaviors via negative reinforcement mechanisms. Decrease in dopamine neurotransmission in the nucleus accumbens and recruitment of corticotropin-releasing factor in the extended amygdala are hypothesized to be implicated in mediating this motivated behavior. Also, a brain stress response system is hypothesized to increase drug craving and contribute to relapse to drug-seeking behavior during the preoccupation and anticipation stage of dependence caused by the exposure to stress characterized as the nonspecific responses to any demands on the body. Acupuncture has proven to be effective for reducing drug addiction and stress-related psychiatric disorders, such as anxiety and depression. Furthermore, acupuncture has been shown to correct reversible brain malfunctions by regulating drug addiction and stress-related neurotransmitters. Accordingly, it seems reasonable to propose that acupuncture attenuates relapse to drug-seeking behavior through inhibition of stress response. In this review, a brief description of stress in relapse to drug-seeking behavior and the effects of acupuncture were presented.
Collapse
|
17
|
Zsilla G, Hegyi DE, Baranyi M, Vizi ES. 3,4-Methylenedioxymethamphetamine, mephedrone, and β-phenylethylamine release dopamine from the cytoplasm by means of transporters and keep the concentration high and constant by blocking reuptake. Eur J Pharmacol 2018; 837:72-80. [DOI: 10.1016/j.ejphar.2018.08.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/24/2018] [Accepted: 08/29/2018] [Indexed: 02/02/2023]
|
18
|
Role of the endocannabinoid system in drug addiction. Biochem Pharmacol 2018; 157:108-121. [PMID: 30217570 DOI: 10.1016/j.bcp.2018.09.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/10/2018] [Indexed: 12/20/2022]
Abstract
Drug addiction is a chronic relapsing disorder that produces a dramaticglobal health burden worldwide. Not effective treatment of drug addiction is currently available probably due to the difficulties to find an appropriate target to manage this complex disease raising the needs for further identification of novel therapeutic approaches. The endocannabinoid system has been found to play a crucial role in the neurobiological substrate underlying drug addiction. Endocannabinoids and cannabinoid receptors are widely expressed in the main areas of the mesocorticolimbic system that participate in the initiation and maintenance of drug consumption and in the development of compulsion and loss of behavioral control occurring during drug addiction. The identification of the important role played by CB1 cannabinoid receptors in drug addiction encouraged the possible used of an early commercialized CB1 receptor antagonist for treating drug addiction. However, the incidence of serious psychiatric adverse events leaded to the sudden withdrawal from the market of this CB1 antagonist and all the research programs developed by pharmaceutical companies to obtain new CB1 antagonists were stopped. Currently, new research strategies are under development to target the endocannabinoid system for drug addiction avoiding these side effects, which include allosteric negative modulators of CB1 receptors and compounds targeting CB2 receptors. Recent studies showing the potential role of CB2 receptors in the addictive properties of different drugs of abuse have open a promising research opportunity to develop novel possible therapeutic approaches.
Collapse
|
19
|
Krolick KN, Zhu Q, Shi H. Effects of Estrogens on Central Nervous System Neurotransmission: Implications for Sex Differences in Mental Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 160:105-171. [PMID: 30470289 PMCID: PMC6737530 DOI: 10.1016/bs.pmbts.2018.07.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nearly one of every five US individuals aged 12 years old or older lives with certain types of mental disorders. Men are more likely to use various types of substances, while women tend to be more susceptible to mood disorders, addiction, and eating disorders, all of which are risks associated with suicidal attempts. Fundamental sex differences exist in multiple aspects of the functions and activities of neurotransmitter-mediated neural circuits in the central nervous system (CNS). Dysregulation of these neural circuits leads to various types of mental disorders. The potential mechanisms of sex differences in the CNS neural circuitry regulating mood, reward, and motivation are only beginning to be understood, although they have been largely attributed to the effects of sex hormones on CNS neurotransmission pathways. Understanding this topic is important for developing prevention and treatment of mental disorders that should be tailored differently for men and women. Studies using animal models have provided important insights into pathogenesis, mechanisms, and new therapeutic approaches of human diseases, but some concerns remain to be addressed. The purpose of this chapter is to integrate human and animal studies involving the effects of the sex hormones, estrogens, on CNS neurotransmission, reward processing, and associated mental disorders. We provide an overview of existing evidence for the physiological, behavioral, cellular, and molecular actions of estrogens in the context of controlling neurotransmission in the CNS circuits regulating mood, reward, and motivation and discuss related pathology that leads to mental disorders.
Collapse
Affiliation(s)
- Kristen N Krolick
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States
| | - Qi Zhu
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States
| | - Haifei Shi
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States; Cellular, Molecular and Structural Biology, Miami University, Oxford, OH, United States.
| |
Collapse
|
20
|
Wenzel JM, Cheer JF. Endocannabinoid Regulation of Reward and Reinforcement through Interaction with Dopamine and Endogenous Opioid Signaling. Neuropsychopharmacology 2018; 43:103-115. [PMID: 28653666 PMCID: PMC5719091 DOI: 10.1038/npp.2017.126] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/01/2017] [Accepted: 06/08/2017] [Indexed: 12/11/2022]
Abstract
The endocannabinoid system (eCB) is implicated in the mediation of both reward and reinforcement. This is evidenced by the ability of exogenous cannabinoid drugs to produce hedonia and maintain self-administration in both human and animal subjects. eCBs similarly facilitate behaviors motivated by reward through interaction with the mesolimbic dopamine (DA) and endogenous opioid systems. Indeed, eCB signaling in the ventral tegmental area stimulates activation of midbrain DA cells and promotes DA release in terminal regions such as the nucleus accumbens (NAc). DA transmission mediates several aspects of reinforced behavior, such as motivation, incentive salience, and cost-benefit calculations. However, much research suggests that endogenous opioid signaling underlies the hedonic aspects of reward. eCBs and their receptors functionally interact with opioid systems within the NAc to support reward, most likely through augmenting DA release. This review explores the interaction of these systems as it relates to reward and reinforcement and examines current literature regarding their role in food reward.
Collapse
Affiliation(s)
- J M Wenzel
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA,Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA,Department of Anatomy and Neurobiology, Department of Psychiatry, Graduate Program in Neuroscience, University of Maryland School of Medicine, HSF I, Room 280J, 20 Penn Street, Baltimore, MD 21201, USA, Tel: +1 410 7060112, Fax: +1 410 7062512, E-mail:
| |
Collapse
|
21
|
Jafari-Sabet M, Karimi AM. Cross state-dependency of learning between arachidonylcyclopropylamide (ACPA) and muscimol in the mouse dorsal hippocampus. Pharmacol Biochem Behav 2017; 163:66-73. [PMID: 29032058 DOI: 10.1016/j.pbb.2017.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 10/09/2017] [Accepted: 10/11/2017] [Indexed: 12/29/2022]
Abstract
The aim of the present study was to examine cross state-dependent learning between ACPA (a selective cannabinoid CB1 receptor agonist) and muscimol (a selective GABAA receptor agonist) in the step-down inhibitory avoidance learning task. The dorsal hippocampal CA1 regions of adult male NMRI mice were bilaterally cannulated, and all drugs were microinjected into the intended sites of injection. Post-training and/or pre-test administration of ACPA (1 and 2ng/mouse) dose-dependently induced amnesia. Pre-test microinjection of the same doses of ACPA reversed the post-training ACPA-induced amnesia. This event has been named ACPA state-dependent learning (SDL). Post-training and/or pre-test microinjection of muscimol (0.05 and 0.1μg/mouse) dose-dependently induced amnesia. Pre-test administration of the same doses of muscimol reversed the post-training muscimol-induced amnesia, suggesting muscimol SDL. The amnesia induced by post-training administration of ACPA was reversed by pre-test administration of muscimol (0.05 and 0.1μg/mouse). Furthermore, the pre-test microinjection of muscimol (0.025 and 0.05μg/mouse) with an ineffective dose of ACPA (0.5ng/mouse) significantly restored memory retrieval and induced ACPA SDL. In another series of experiments, the amnesia induced by post-training administration of muscimol was reversed by pre-test administration of ACPA (1 and 2ng/mouse). Moreover, pre-test microinjection of ACPA (0.5 and 1ng/mouse) with an ineffective dose of muscimol (0.025μg/mouse) significantly restored memory retrieval and induced muscimol SDL. It is important to note that pre-test intra-CA1 injection of a selective GABAA receptor antagonist, bicuculline (0.125 and 0.25μg/mouse), 5min before the administration of muscimol (0.1μg/mouse) or ACPA (2ng/mouse) dose-dependently inhibited muscimol- and ACPA-induced SDL, respectively. Pre-test intra-CA1 administration of bicuculline (0.0625, 0.125 and 0.25μg/mouse) by itself did not affect memory retention. In conclusion, the data strongly revealed a cross SDL among ACPA and muscimol in the dorsal hippocampal CA1 regions.
Collapse
Affiliation(s)
- Majid Jafari-Sabet
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Amir-Mohammad Karimi
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Rakovska A, Baranyi M, Windisch K, Petkova-Kirova P, Gagov H, Kalfin R. Neurochemical evidence that cocaine- and amphetamine-regulated transcript (CART) 55-102 peptide modulates the dopaminergic reward system by decreasing the dopamine release in the mouse nucleus accumbens. Brain Res Bull 2017; 134:246-252. [PMID: 28802898 DOI: 10.1016/j.brainresbull.2017.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/05/2017] [Accepted: 08/07/2017] [Indexed: 01/11/2023]
Abstract
CART (Cocaine- and Amphetamine-Regulated Transcript) peptide is a neurotransmitter naturally occurring in the CNS and found mostly in nucleus accumbens, ventrotegmental area, ventral pallidum, amygdalae and striatum, brain regions associated with drug addiction. In the nucleus accumbens, known for its significant role in motivation, pleasure, reward and reinforcement learning, CART peptide inhibits cocaine and amphetamine-induced dopamine-mediated increases in locomotor activity and behavior, suggesting a CART peptide interaction with the dopaminergic system. Thus in the present study, we examined the effect of CART (55-102) peptide on the basal, electrical field stimulation-evoked (EFS-evoked) (30V, 2Hz, 120 shocks) and returning basal dopamine (DA) release and on the release of the DA metabolites 3,4-dihydroxyphenyl acetaldehyde (DOPAL), 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), 3,4-dihydroxyphenylethanol (DOPET), 3-methoxytyramine (3-MT) as well as on norepinephrine (NE) and dopamine-o-quinone (Daq) in isolated mouse nucleus accumbens, in a preparation, in which any CART peptide effects on the dendrites or soma of ventral tegmental projection neurons have been excluded. We further extended our study to assess the effect of CART (55-102) peptide on basal cocaine-induced release of dopamine and its metabolites DOPAL, DOPAC, HVA, DOPET and 3-MT as well as on NE and Daq. To analyze the amount of [3H]dopamine, dopamine metabolites, Daq and NE in the nucleus accumbens superfusate, a high-pressure liquid chromatography (HPLC), coupled with electrochemical, UV and radiochemical detections was used. CART (55-102) peptide, 0.1μM, added alone, exerted: (i) a significant decrease in the basal and EFS-evoked levels of extracellular dopamine (ii) a significant increase in the EFS-evoked and returning basal levels of the dopamine metabolites DOPAC and HVA, major products of dopamine degradation and (iii) a significant decrease in the returning basal levels of DOPET. At the same concentration, 0.1μM, CART (55-102) peptide did not have any effect on the release of noradrenaline. In the presence of CART (55-102) peptide, 0.1μM, the effect of cocaine, 30μM, on the basal dopamine release was inhibited and the effect on the basal DOPAC release substantially increased. To our knowledge, our findings are the first to show direct neurochemical evidence that CART (55-102) peptide plays a neuromodulatory role on the dopaminergic reward system by decreasing dopamine in the mouse nucleus accumbens and by attenuating cocaine-induced effects on dopamine release.
Collapse
Affiliation(s)
- Angelina Rakovska
- Lab. "Neuropeptides", Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113, Sofia, Bulgaria.
| | - Maria Baranyi
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u. 43, H-1083, Budapest, Hungary
| | - Katalin Windisch
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u. 43, H-1083, Budapest, Hungary
| | - Polina Petkova-Kirova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. bl. 21, 1113, Sofia, Bulgaria
| | - Hristo Gagov
- Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Str. 8, 1164, Sofia, Bulgaria
| | - Reni Kalfin
- Lab. "Neuropeptides", Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113, Sofia, Bulgaria
| |
Collapse
|
23
|
Fouyssac M, Everitt BJ, Belin D. Cellular basis of the intrastriatal functional shifts that underlie the development of habits: relevance for drug addiction. Curr Opin Behav Sci 2017. [DOI: 10.1016/j.cobeha.2016.11.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Torres JAKL, Rosales RL. Nonmotor Symptoms in Dystonia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 134:1335-1371. [DOI: 10.1016/bs.irn.2017.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
25
|
Peterson BM, Martinez LA, Meisel RL, Mermelstein PG. Estradiol impacts the endocannabinoid system in female rats to influence behavioral and structural responses to cocaine. Neuropharmacology 2016; 110:118-124. [PMID: 27266915 PMCID: PMC5028287 DOI: 10.1016/j.neuropharm.2016.06.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/31/2016] [Accepted: 06/02/2016] [Indexed: 12/31/2022]
Abstract
Compared with men, women show enhanced responses to drugs of abuse, and consequently are thought to be more vulnerable to addiction. The ovarian hormone estradiol has emerged as a key facilitator in the heightened development of addiction in females. These actions of estradiol appear mediated by estrogen receptor (ER) activation of metabotropic glutamate receptor type 5 (mGluR5). However, the downstream effectors of this ER/mGluR5 signaling pathway are unknown. Here we investigate whether cannabinoid 1 receptor (CB1R) activation is a part of the mechanism whereby estradiol influences behavioral and synaptic correlates of addiction. Following repeated cocaine administration, estradiol-treated ovariectomized rats exhibited both sensitized locomotor responses and decreases in the dendritic spine density of nucleus accumbens core medium-spiny neurons in comparison to oil-treated controls. Both effects of estradiol were blocked by AM251, a CB1R inverse agonist. These results indicate that part of the signaling mechanism through which estradiol impacts behavioral and synaptic correlates of addiction in female rats requires activation of CB1Rs.
Collapse
Affiliation(s)
- Brittni M Peterson
- Graduate Program in Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA; Department of Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Luis A Martinez
- Department of Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Robert L Meisel
- Graduate Program in Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA; Department of Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Paul G Mermelstein
- Graduate Program in Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA; Department of Neuroscience, 6-145 Jackson Hall, 321 Church Street SE, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
26
|
Guegan T, Cebrià JP, Maldonado R, Martin M. Morphine-induced locomotor sensitization produces structural plasticity in the mesocorticolimbic system dependent on CB1-R activity. Addict Biol 2016; 21:1113-1126. [PMID: 26179931 DOI: 10.1111/adb.12281] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Changes in structural plasticity produced by the chronic exposure to drugs of abuse, such as alterations in dendritic spine densities, participate in the development of maladaptive learning processes leading to drug addiction. Understanding the neurobiological mechanisms involved in these aberrant changes is crucial to clarify the neurobiological substrate of addiction. Drug-induced locomotor sensitization has been widely accepted as a useful animal model to study these mechanisms related to drug addiction. We have evaluated the changes in structural plasticity in the mesocorticolimbic system involved in morphine-induced locomotor sensitization. The role of the cannabinoid receptor type 1 (CB1-R) in these neuroplastic alterations has also been studied using CB1-R-deficient (CB1-R KO) mice. Structural plasticity changes promoted by morphine are a highly dynamic phenomenon that evolves during the entire time course of the behavioral sensitization in wild-type (WT) animals. The different phases of the sensitization process were related to specific changes in connectivity between neurons revealed by modifications in dendritic spines in specific areas of the mesocorticolimbic system. Moreover, the lack of morphine-induced locomotor sensitization in CB1-R KO mice was accompanied by abnormal alterations in structural plasticity in the same mesocorticolimbic areas. These specific structural plasticity changes mediated by CB1-R activity seem necessary for the normal progression of morphine-induced locomotor sensitization and could play a critical role in the addictive process.
Collapse
Affiliation(s)
- Thomas Guegan
- Laboratory of Neuropharmacology; Parc de Recerca Biomèdica de Barcelona/Universitat Pompeu Fabra; Spain
| | - Joan Pau Cebrià
- Laboratory of Neuropharmacology; Parc de Recerca Biomèdica de Barcelona/Universitat Pompeu Fabra; Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology; Parc de Recerca Biomèdica de Barcelona/Universitat Pompeu Fabra; Spain
| | - Miquel Martin
- Laboratory of Neuropharmacology; Parc de Recerca Biomèdica de Barcelona/Universitat Pompeu Fabra; Spain
| |
Collapse
|
27
|
Estradiol Facilitation of Cocaine Self-Administration in Female Rats Requires Activation of mGluR5. eNeuro 2016; 3:eN-NWR-0140-16. [PMID: 27822496 PMCID: PMC5079229 DOI: 10.1523/eneuro.0140-16.2016] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 10/07/2016] [Accepted: 10/07/2016] [Indexed: 02/06/2023] Open
Abstract
In comparison to men, women initiate drug use at earlier ages and progress from initial use to addiction more rapidly. This heightened intake and vulnerability to drugs of abuse is regulated in part by estradiol, although the signaling mechanisms by which this occurs are not well understood. Recent findings indicate that within the nucleus accumbens core, estradiol induces structural plasticity via membrane-localized estrogen receptor α, functionally coupled to metabotropic glutamate receptor subtype 5 (mGluR5). Hence, we sought to determine whether mGluR5 activation was essential for estradiol-mediated enhancement of cocaine self-administration. Ovariectomized (OVX) female rats were allowed to freely self-administer cocaine under extended access conditions (6 h/d) for 10 consecutive days. The mGluR5 antagonist 2-methyl-6-(phenylethynyl)pyridine hydrochloride (MPEP) or vehicle was administered before estradiol (or oil), on a 2 d on/2 d off schedule throughout the extended access period. MPEP treatment prevented the estradiol-dependent enhancement of cocaine self-administration in OVX females. In a separate experiment, potentiation of mGluR5 function with the positive allosteric modulator 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (in the absence of estradiol treatment) failed to increase cocaine self-administration. These data suggest that mGluR5 activation is necessary for estradiol-mediated enhancement of responses to cocaine, but that direct mGluR5 activation is insufficient to mimic the female response to estradiol. Building on previous studies in male animals, these findings further highlight the therapeutic potential of mGluR5 antagonism in the treatment of addiction and suggest that there may be added therapeutic benefit in females.
Collapse
|
28
|
Sex differences in alcohol consumption and alterations in nucleus accumbens endocannabinoid mRNA in alcohol-dependent rats. Neuroscience 2016; 335:195-206. [DOI: 10.1016/j.neuroscience.2016.08.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 11/22/2022]
|
29
|
Nasehi M, Kamali-Dolatabadi L, Torabi-Nami M, Zarrindast MR. Possible involvement of the CA1 GABAA receptors upon acquisition and expression of the ACPA-induced place preference in mice. Physiol Behav 2016; 161:155-165. [DOI: 10.1016/j.physbeh.2016.04.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 04/12/2016] [Accepted: 04/12/2016] [Indexed: 12/12/2022]
|
30
|
Pinheiro BS, Lemos C, Neutzling Kaufmann F, Marques JM, da Silva-Santos CS, Carvalho E, Mackie K, Rodrigues RJ, Cunha RA, Köfalvi A. Hierarchical glucocorticoid-endocannabinoid interplay regulates the activation of the nucleus accumbens by insulin. Brain Res Bull 2016; 124:222-30. [PMID: 27208730 DOI: 10.1016/j.brainresbull.2016.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 12/28/2022]
Abstract
Here we asked if insulin activation of the nucleus accumbens in vitro is reflected by an increase in (3)H-deoxyglucose ([(3)H]DG) uptake, thus subserving a new model to study molecular mechanisms of central insulin actions. Additionally, we investigated the dependence of this insulin effect on endocannabinoids and corticosteroids, two major culprits in insulin resistance. We found that in acute accumbal slices, insulin (3 and 300nM but not at 0.3nM) produced an increase in [(3)H]DG uptake. The synthetic cannabinoid agonist, WIN55212-2 (500nM) and the glucocorticoid dexamethasone (10μM), impaired insulin (300nM) action on [(3)H]DG uptake. The glucocorticoid receptor (GcR) antagonist, mifepristone (10μM) prevented dexamethasone from inhibiting insulin's action. Strikingly, this anti-insulin action of dexamethasone was also blocked by two CB1 cannabinoid receptor (CB1R) antagonists, O-2050 (500nM) and SR141716A (500nM), as well as by tetrahydrolipstatin (10μM), an inhibitor of diacylglycerol lipases-the enzymes responsible for the synthesis of the endocannabinoid, 2-arachidonoyl-glycerol (2-AG). On the other hand, the blockade of the post-synaptic 2-AG metabolizing enzymes, α,β-serine hydrolase domain 6/12 by WWL70 (1μM) also prevented the action of insulin, probably via increasing endogenous 2-AG tone. Additionally, an anti-insulin receptor (InsR) antibody immunoprecipitated CB1Rs from accumbal homogenates, indicating a physical complexing of CB1Rs with InsRs that supports their functional interaction. Altogether, insulin stimulates glucose uptake in the nucleus accumbens. Accumbal GcR activation triggers the synthesis of 2-AG that in turn binds to the known CB1R-InsR heteromer, thus impeding insulin signaling.
Collapse
Affiliation(s)
- Bárbara S Pinheiro
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Cristina Lemos
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | - Joana M Marques
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Carla S da Silva-Santos
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Eugénia Carvalho
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - Ricardo J Rodrigues
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; FMUC, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Attila Köfalvi
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal.
| |
Collapse
|
31
|
Moreira FA, Jupp B, Belin D, Dalley JW. Endocannabinoids and striatal function: implications for addiction-related behaviours. Behav Pharmacol 2015; 26:59-72. [PMID: 25369747 PMCID: PMC5398317 DOI: 10.1097/fbp.0000000000000109] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 09/26/2014] [Indexed: 12/24/2022]
Abstract
Since the identification and cloning of the major cannabinoid receptor expressed in the brain almost 25 years ago research has highlighted the potential of drugs that target the endocannabinoid system for treating addiction. The endocannabinoids, anandamide and 2-arachidonoyl glycerol, are lipid-derived metabolites found in abundance in the basal ganglia and other brain areas innervated by the mesocorticolimbic dopamine systems. Cannabinoid CB1 receptor antagonists/inverse agonists reduce reinstatement of responding for cocaine, alcohol and opiates in rodents. However, compounds acting on the endocannabinoid system may have broader application in treating drug addiction by ameliorating associated traits and symptoms such as impulsivity and anxiety that perpetuate drug use and interfere with rehabilitation. As a trait, impulsivity is known to predispose to addiction and facilitate the emergence of addiction to stimulant drugs. In contrast, anxiety and elevated stress responses accompany extended drug use and may underlie the persistence of drug intake in dependent individuals. In this article we integrate and discuss recent findings in rodents showing selective pharmacological modulation of impulsivity and anxiety by cannabinoid agents. We highlight the potential of selective inhibitors of endocannabinoid metabolism, directed at fatty acid amide hydrolase and monoacylglycerol lipase, to reduce anxiety and stress responses, and discuss novel mechanisms underlying the modulation of the endocannabinoid system, including the attenuation of impulsivity, anxiety, and drug reward by selective CB2 receptor agonists.
Collapse
Affiliation(s)
- Fabricio A. Moreira
- Department of Pharmacology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Departments of Psychology
| | | | | | - Jeffrey W. Dalley
- Departments of Psychology
- Department of Psychiatry, Addenbrookes’s Hospital University of Cambridge, Cambridge, UK
| |
Collapse
|
32
|
The microinjection of a cannabinoid agonist into the accumbens shell induces anxiogenesis in the elevated plus-maze. Pharmacol Biochem Behav 2014; 124:160-6. [PMID: 24887448 DOI: 10.1016/j.pbb.2014.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/19/2014] [Accepted: 05/22/2014] [Indexed: 11/23/2022]
Abstract
This study investigated the effect of a cannabinoid agonist injected into the shell region of the nucleus accumbens (nAcb shell) on anxiety-related behaviors. The animals (male Wistar rats) were unilaterally microinjected with either ACEA (arachidonyl-2'-chloroethylamide a CB1 receptor agonist) at doses of 0.005, 0.05 or 0.5 pmol, or vehicle (ethanol 0.04% in saline 0.9%) and submitted to the elevated plus-maze (EPM), a pre-clinical test of anxiety. The data showed that rats microinjected with ACEA (0.05 pmol/0.2 μl) into the nAcb shell exhibited decreased % open arm time and open arm entries in comparison with the control group, which is compatible with an anxiogenic-like effect. To rule out the hypothesis that spread of the drug into the ventricle was responsible for the observed anxiogenic effect, 0.05 pmol ACEA was injected into the lateral ventricle and shown not to alter the responses representative of fear/anxiety and locomotion. The locomotor activity was not changed at the dose of 0.05 pmol ACEA microinjected into the nAcb shell. The present data suggest that activation of cannabinoid receptors in the nAcb shell may modulate fear/anxiety in the EPM.
Collapse
|
33
|
Tree KC, Scotto di Perretolo M, Peyronnet J, Cayetanot F. In utero cannabinoid exposure alters breathing and the response to hypoxia in newborn mice. Eur J Neurosci 2014; 40:2196-204. [PMID: 24717006 DOI: 10.1111/ejn.12588] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 01/21/2014] [Accepted: 03/13/2014] [Indexed: 11/26/2022]
Abstract
Cannabis is one of the most commonly used recreational drugs at ages highly correlated with potential pregnancy. Endocannabinoid signalling regulates important stages of neuronal development. When cannabinoid receptors, which are widely distributed through the nervous system, are activated by exogenous cannabinoids, breathing in adult rats is depressed. Here, we show that, in newborn mice, endocannabinoids, through the activation of cannabinoid receptor type 1 (CB1 R), participate in the modulation of respiration and its control. Blocking CB1 Rs at birth suppressed the brake exerted by endocannabinoids on ventilation in basal and in hypoxic conditions. The number of apnoeas and their duration were also minimized by activation of CB1 Rs in normoxic and in hypoxic conditions. However, prenatal cannabis intoxication, caused by a daily injection of WIN55,212-2, in pregnant mice durably modified respiration of the offspring, as shown by hyperventilation in basal conditions, an altered chemoreflex in response to hypoxia, and longer apnoeas. When CB1 Rs were blocked in WIN55,212-2 treated newborns, persistent hyperventilation was still observed, which could partly be explained by a perturbation of the central respiratory network. In fact, in vitro medullary preparations from WIN55,212-2 treated pups, free of peripheral or of supramedullary structures, showed an altered fictive breathing frequency. In conclusion, the endocannabinoid pathway at birth seems to modulate breathing and protect the newborn against apnoeas. However, when exposed prenatally to an excess of cannabinoid, the breathing neuronal network in development seems to be modified, probably rendering the newborn more vulnerable in the face of an unstable environment.
Collapse
Affiliation(s)
- Keda C Tree
- Institut de Neurosciences de la Timone UMR 7289, Aix Marseille Université, CNRS, Marseille, France
| | | | | | | |
Collapse
|
34
|
Matsunaga M, Isowa T, Yamakawa K, Fukuyama S, Shinoda J, Yamada J, Ohira H. Genetic variations in the human cannabinoid receptor gene are associated with happiness. PLoS One 2014; 9:e93771. [PMID: 24690898 PMCID: PMC3972248 DOI: 10.1371/journal.pone.0093771] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 03/07/2014] [Indexed: 01/15/2023] Open
Abstract
Happiness has been viewed as a temporary emotional state (e.g., pleasure) and a relatively stable state of being happy (subjective happiness level). As previous studies demonstrated that individuals with high subjective happiness level rated their current affective states more positively when they experience positive events, these two aspects of happiness are interrelated. According to a recent neuroimaging study, the cytosine to thymine single-nucleotide polymorphism of the human cannabinoid receptor 1 gene is associated with sensitivity to positive emotional stimuli. Thus, we hypothesized that our genetic traits, such as the human cannabinoid receptor 1 genotypes, are closely related to the two aspects of happiness. In Experiment 1, 198 healthy volunteers were used to compare the subjective happiness level between cytosine allele carriers and thymine-thymine carriers of the human cannabinoid receptor 1 gene. In Experiment 2, we used positron emission tomography with 20 healthy participants to compare the brain responses to positive emotional stimuli of cytosine allele carriers to that of thymine-thymine carriers. Compared to thymine-thymine carriers, cytosine allele carriers have a higher subjective happiness level. Regression analysis indicated that the cytosine allele is significantly associated with subjective happiness level. The positive mood after watching a positive film was significantly higher for the cytosine allele carriers compared to the thymine-thymine carriers. Positive emotion-related brain region such as the medial prefrontal cortex was significantly activated when the cytosine allele carriers watched the positive film compared to the thymine-thymine carriers. Thus, the human cannabinoid receptor 1 genotypes are closely related to two aspects of happiness. Compared to thymine-thymine carriers, the cytosine allele carriers of the human cannabinoid receptor 1 gene, who are sensitive to positive emotional stimuli, exhibited greater magnitude positive emotions when they experienced positive events and had a higher subjective happiness level.
Collapse
Affiliation(s)
- Masahiro Matsunaga
- Department of Health and Psychosocial Medicine, Aichi Medical University School of Medicine, Aichi, Japan
- * E-mail:
| | - Tokiko Isowa
- Department of Gerontological Nursing, Mie University Faculty of Medicine, Mie, Japan
| | - Kaori Yamakawa
- Department of Psychology, Graduate School of Environmental Studies, Nagoya University, Aichi, Japan
| | - Seisuke Fukuyama
- Chubu Medical Center for Prolonged Traumatic Brain Dysfunction, Kizawa Memorial Hospital, Gifu, Japan
| | - Jun Shinoda
- Chubu Medical Center for Prolonged Traumatic Brain Dysfunction, Kizawa Memorial Hospital, Gifu, Japan
| | - Jitsuhiro Yamada
- Chubu Medical Center for Prolonged Traumatic Brain Dysfunction, Kizawa Memorial Hospital, Gifu, Japan
| | - Hideki Ohira
- Department of Psychology, Graduate School of Environmental Studies, Nagoya University, Aichi, Japan
| |
Collapse
|
35
|
Tan H, Ahmad T, Loureiro M, Zunder J, Laviolette SR. The role of cannabinoid transmission in emotional memory formation: implications for addiction and schizophrenia. Front Psychiatry 2014; 5:73. [PMID: 25071606 PMCID: PMC4074769 DOI: 10.3389/fpsyt.2014.00073] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 06/13/2014] [Indexed: 11/26/2022] Open
Abstract
Emerging evidence from both basic and clinical research demonstrates an important role for endocannabinoid (ECB) signaling in the processing of emotionally salient information, learning, and memory. Cannabinoid transmission within neural circuits involved in emotional processing has been shown to modulate the acquisition, recall, and extinction of emotionally salient memories and importantly, can strongly modulate the emotional salience of incoming sensory information. Two neural regions in particular, the medial prefrontal cortex (PFC) and the basolateral nucleus of the amygdala (BLA), play important roles in emotional regulation and contain high levels of cannabinoid receptors. Furthermore, both regions show profound abnormalities in neuropsychiatric disorders such as addiction and schizophrenia. Considerable evidence has demonstrated that cannabinoid transmission functionally interacts with dopamine (DA), a neurotransmitter system that is of exceptional importance for both addictive behaviors and the neuropsychopathology of disorders like schizophrenia. Research in our laboratory has focused on how cannabinoid transmission both within and extrinsic to the mesolimbic DA system, including the BLA → mPFC circuitry, can modulate both rewarding and aversive emotional information. In this review, we will summarize clinical and basic neuroscience research demonstrating the importance of cannabinoid signaling within this neural circuitry. In particular, evidence will be reviewed emphasizing the importance of cannabinoid signaling within the BLA → mPFC circuitry in the context of emotional salience processing, memory formation and memory-related plasticity. We propose that aberrant states of hyper or hypoactive ECB signaling within the amygdala-prefrontal cortical circuit may lead to dysregulation of mesocorticolimbic DA transmission controlling the processing of emotionally salient information. These disturbances may in turn lead to emotional processing, learning, and memory abnormalities related to various neuropsychiatric disorders, including addiction and schizophrenia-related psychoses.
Collapse
Affiliation(s)
- Huibing Tan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London, ON , Canada
| | - Tasha Ahmad
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London, ON , Canada
| | - Michael Loureiro
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London, ON , Canada
| | - Jordan Zunder
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London, ON , Canada
| | - Steven R Laviolette
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London, ON , Canada ; Department of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario , London, ON , Canada ; Department of Psychology, Schulich School of Medicine and Dentistry, University of Western Ontario , London, ON , Canada
| |
Collapse
|
36
|
Ahmad T, Lauzon NM, de Jaeger X, Laviolette SR. Cannabinoid transmission in the prelimbic cortex bidirectionally controls opiate reward and aversion signaling through dissociable kappa versus μ-opiate receptor dependent mechanisms. J Neurosci 2013; 33:15642-51. [PMID: 24068830 PMCID: PMC6618460 DOI: 10.1523/jneurosci.1686-13.2013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 08/12/2013] [Accepted: 08/27/2013] [Indexed: 11/21/2022] Open
Abstract
Cannabinoid, dopamine (DA), and opiate receptor pathways play integrative roles in emotional learning, associative memory, and sensory perception. Modulation of cannabinoid CB1 receptor transmission within the medial prefrontal cortex (mPFC) regulates the emotional valence of both rewarding and aversive experiences. Furthermore, CB1 receptor substrates functionally interact with opiate-related motivational processing circuits, particularly in the context of reward-related learning and memory. Considerable evidence demonstrates functional interactions between CB1 and DA signaling pathways during the processing of motivationally salient information. However, the role of mPFC CB1 receptor transmission in the modulation of behavioral opiate-reward processing is not currently known. Using an unbiased conditioned place preference paradigm with rats, we examined the role of intra-mPFC CB1 transmission during opiate reward learning. We report that activation or inhibition of CB1 transmission within the prelimbic cortical (PLC) division of the mPFC bidirectionally regulates the motivational valence of opiates; whereas CB1 activation switched morphine reward signaling into an aversive stimulus, blockade of CB1 transmission potentiated the rewarding properties of normally sub-reward threshold conditioning doses of morphine. Both of these effects were dependent upon DA transmission as systemic blockade of DAergic transmission prevented CB1-dependent modulation of morphine reward and aversion behaviors. We further report that CB1-mediated intra-PLC opiate motivational signaling is mediated through a μ-opiate receptor-dependent reward pathway, or a κ-opiate receptor-dependent aversion pathway, directly within the ventral tegmental area. Our results provide evidence for a novel CB1-mediated motivational valence switching mechanism within the PLC, controlling dissociable subcortical reward and aversion pathways.
Collapse
MESH Headings
- Animals
- Conditioning, Classical
- Limbic System/metabolism
- Limbic System/physiology
- Male
- Motivation
- Neurotransmitter Agents/pharmacology
- Prefrontal Cortex/metabolism
- Prefrontal Cortex/physiology
- Rats
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Reward
- Synaptic Transmission
- Ventral Tegmental Area/metabolism
- Ventral Tegmental Area/physiology
Collapse
Affiliation(s)
| | | | | | - Steven R. Laviolette
- Departments of Anatomy and Cell Biology
- Psychiatry, and
- Psychology, The Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N5Y 5T8
| |
Collapse
|
37
|
Kleijn J, Wiskerke J, Cremers T, Schoffelmeer A, Westerink B, Pattij T. Effects of amphetamine on dopamine release in the rat nucleus accumbens shell region depend on cannabinoid CB1 receptor activation. Neurochem Int 2012; 60:791-8. [DOI: 10.1016/j.neuint.2012.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Revised: 02/29/2012] [Accepted: 03/01/2012] [Indexed: 10/28/2022]
|
38
|
Farkas S, Nagy K, Palkovits M, Kovács GG, Jia Z, Donohue S, Pike V, Halldin C, Máthé D, Harkany T, Gulyás B, Csiba L. [¹²⁵I]SD-7015 reveals fine modalities of CB₁ cannabinoid receptor density in the prefrontal cortex during progression of Alzheimer's disease. Neurochem Int 2011; 60:286-91. [PMID: 22222721 DOI: 10.1016/j.neuint.2011.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 10/25/2011] [Accepted: 11/08/2011] [Indexed: 02/01/2023]
Abstract
The cannabinoid type-1 receptor (CB₁R) is one of the most abundant members of the G protein-coupled receptor family in the central nervous system. Once activated by their cognate ligands, endocannabinoids, CB₁Rs generally limit the timing of neurotransmitter release at many cortical synapses. Prior studies have indicated the involvement of CB₁R in neurodegeneration and in various neuronal insults, with an emphasis on their neuroprotective role. In the present study we used a novel selective CB₁R radioligand to investigate regional variations in CB₁R ligand binding as a factor of progressive Braak tau pathology in the frontal cortex of Alzheimer's disease (AD) patients. The frontal cortex was chosen for this study due to the high density of CB₁Rs and their well-characterized involvement in the progression of AD. Post-mortem prefrontal cortex samples from AD patients from Braak stages I to VI and controls were subjected to CB₁R autoradiography with [¹²⁵I]SD-7015 as radioligand. Regional concentration of [¹²⁵I]SD-7015, corresponding to, and thereby representing, regional CB₁R densities, were expressed in fM/g_tissue. The results show that CB₁R density inversely correlates with Braak tau pathology with the following tendency: controls <AD Braak stage V-VI <AD Braak stage III-IV <AD Braak stage I-II. Differences were significant between control and AD Braak stage I-II groups, as well as between controls and the AD group comprising all Braak stages. These findings indicate an up-regulation of the tissue binding of the selective CB₁R radioligand [¹²⁵I]SD7015 in human brains, allowing the detection of fine modalities of receptor expression and radioligand binding during the progression of AD.
Collapse
Affiliation(s)
- Szabolcs Farkas
- Department of Neurology, University of Debrecen, H-4012 Debrecen, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Andó RD, Bíró J, Csölle C, Ledent C, Sperlágh B. The inhibitory action of exo- and endocannabinoids on [³H]GABA release are mediated by both CB₁and CB₂receptors in the mouse hippocampus. Neurochem Int 2011; 60:145-52. [PMID: 22133429 DOI: 10.1016/j.neuint.2011.11.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 10/19/2011] [Accepted: 11/15/2011] [Indexed: 11/15/2022]
Abstract
Exogenous and endogenous cannabinoids play an important role in modulating the release of neurotransmitters in hippocampal excitatory and inhibitory networks, thus having profound effect on higher cognitive and emotional functions such as learning and memory. In this study we have studied the effect of cannabinoid agonists on the potassium depolarization-evoked [(3)H]GABA release from hippocampal synaptosomes in the wild-type (WT) and cannabinoid 1 receptor (CB(1)R)-null mutant mice. All tested cannabinoid agonists (WIN55,212-2, CP55,940, HU-210, 2-arachidonoyl-glycerol, 2-AG; delta-9-tetra-hydrocannabinol, THC) inhibited [(3)H]GABA release in WT mice with the following rank order of agonist potency: HU-210>CP55,490>WIN55,212-2>>2-AG>THC. By contrast, 2-AG and THC displayed the greatest efficacy eliciting almost complete inhibition of evoked [(3)H]GABA efflux, whereas the maximal inhibition obtained by HU-210, CP55,490, and WIN55,212-2 were less, eliciting not more than 40% inhibition. The inhibitory effect of WIN55,212-2, THC and 2-AG on evoked [(3)H]GABA efflux was antagonized by the CB(1) receptor inverse agonist AM251 (0.5 μM) in the WT mice. In the CB(1)R knockout mice the inhibitory effects of all three agonists were attenuated. In these mice, AM251 did not antagonize, but further reduced the [(3)H]GABA release in the presence of the synthetic agonist WIN55,212-2. By contrast, the concentration-dependent inhibitory effects of THC and 2-AG were partially antagonized by AM251 in the absence of CB(1) receptors. Finally, the inhibition of evoked [(3)H]GABA efflux by THC and 2-AG was also partially attenuated by AM630 (1 μM), the CB(2) receptor-selective antagonist, both in WT and CB(1) knockout mice. Our data prove the involvement of CB(1) receptors in the effect of exo- and endocannabinoids on GABA efflux from hippocampal nerve terminals. In addition, in the effect of the exocannabinoid THC and the endocannabinoid 2-AG, non-CB(1), probably CB(2)-like receptors are also involved.
Collapse
MESH Headings
- Animals
- Cannabinoid Receptor Modulators/physiology
- Cannabinoids/pharmacology
- Endocannabinoids
- Hippocampus/metabolism
- Hippocampus/physiology
- Male
- Mice
- Mice, Inbred Strains
- Mice, Knockout
- Neural Inhibition/genetics
- Neural Inhibition/physiology
- Presynaptic Terminals/metabolism
- Presynaptic Terminals/physiology
- Receptor, Cannabinoid, CB1/deficiency
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/genetics
- Receptor, Cannabinoid, CB2/metabolism
- Tritium
- gamma-Aminobutyric Acid/metabolism
Collapse
Affiliation(s)
- Rómeó D Andó
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | | | |
Collapse
|
40
|
Seif T, Makriyannis A, Kunos G, Bonci A, Hopf FW. The endocannabinoid 2-arachidonoylglycerol mediates D1 and D2 receptor cooperative enhancement of rat nucleus accumbens core neuron firing. Neuroscience 2011; 193:21-33. [PMID: 21821098 DOI: 10.1016/j.neuroscience.2011.07.055] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 07/14/2011] [Accepted: 07/22/2011] [Indexed: 01/09/2023]
Abstract
Many motivated and addiction-related behaviors are sustained by activity of both dopamine D1- and D2-type receptors (D1Rs and D2Rs) as well as CB1 receptors (CB1Rs) in the nucleus accumbens (NAc). Here, we use in vitro whole-cell patch-clamp electrophysiology to describe an endocannabinoid (eCB)-dopamine receptor interaction in adult rat NAc core neurons. D1R and D2R agonists in combination enhanced firing, with no effect of a D1R or D2R agonist alone. This D1R+D2R-mediated firing increase required CB1Rs, since it was prevented by the CB1R antagonists AM251 and Rimonabant. The D1R+D2R firing increase also required phospholipase C (PLC), the major synthesis pathway for the eCB 2-arachidonoylglycerol (2-AG) and one of several pathways for anandamide. Further, inhibition of 2-AG hydrolysis with the monoglyceride lipase (MGL) inhibitor JZL184 allowed subthreshold levels of D1R+D2R receptor agonists to enhance firing, while inhibition of anandamide hydrolysis with the fatty acid amide hydrolase (FAAH) inhibitors URB597 or AM3506 did not. Filling the postsynaptic neuron with 2-AG enabled subthreshold D1R+D2R agonists to increase firing, and the 2AG+D1R+D2R increase in firing was prevented by a CB1R antagonist. Also, the metabotropic glutamate receptor 5 (mGluR5) blocker MPEP prevented the ability of JZL184 to promote subthreshold D1R+D2R enhancement of firing, while the 2-AG+D1R+D2R increase in firing was not prevented by the mGluR5 blocker, suggesting that mGluR5s acted upstream of 2-AG production. Thus, our results taken together are consistent with the hypothesis that NAc core eCBs mediate dopamine receptor (DAR) enhancement of firing, perhaps providing a cellular mechanism underlying the central role of NAc core D1Rs, D2Rs, CB1Rs, and mGluR5s during many drug-seeking behaviors.
Collapse
Affiliation(s)
- T Seif
- Ernest Gallo Clinic and Research Center, University of California, San Francisco, Department of Neurology, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA
| | | | | | | | | |
Collapse
|
41
|
Cannabinoid receptor blockade reduces the opportunity cost at which rats maintain operant performance for rewarding brain stimulation. J Neurosci 2011; 31:5426-35. [PMID: 21471378 DOI: 10.1523/jneurosci.0079-11.2011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
There is ample evidence that blockade of CB(1) receptors reduces reward seeking. However, the reported effects of CB(1) blockade on performance for rewarding electrical brain stimulation stand out as an exception. By applying a novel method for conceptualizing and measuring reward seeking, we show that AM-251, a CB(1) receptor antagonist, does indeed decrease performance for rewarding electrical stimulation of the medial forebrain bundle in rats. Reward seeking depends on multiple sets of variables, including the intensity of the reward, its cost, and the value of competing rewards. In turn, reward intensity depends both on the sensitivity and gain of brain reward circuitry. We show that drug-induced changes in sensitivity cannot account for the suppressive effect of AM-251 on reward seeking. Therefore, the role of CB(1) receptors must be sought among the remaining determinants of performance. Our analysis provides an explanation of the inconsistencies between prior reports, which likely arose from the following: (1) the averaging of data across subjects showing heterogeneous effects and (2) the use of methods that cannot distinguish between the different determinants of reward pursuit. By means of microdialysis, we demonstrate that blockade of CB(1) receptors attenuates nucleus accumbens dopamine release in response to rewarding medial forebrain bundle stimulation, and we propose that this action is responsible for the ability of the drug to decrease performance for the electrical reward.
Collapse
|
42
|
Cannarsa R, Carretta D, Lattanzio F, Candeletti S, Romualdi P. ∆9-Tetrahydrocannabinol Decreases NOP Receptor Density and mRNA Levels in Human SH-SY5Y Cells. J Mol Neurosci 2011; 46:285-92. [DOI: 10.1007/s12031-011-9552-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 05/09/2011] [Indexed: 11/27/2022]
|
43
|
Zarrindast MR, Mahboobi S, Sadat-Shirazi MS, Ahmadi S. Anxiolytic-like effect induced by the cannabinoid CB1 receptor agonist, arachydonilcyclopropylamide (ACPA), in the rat amygdala is mediated through the D1 and D2 dopaminergic systems. J Psychopharmacol 2011; 25:131-40. [PMID: 20685770 DOI: 10.1177/0269881110376688] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the present study the influence of the dopaminergic system(s) of the amygdala on the anxiolytic-like effect of the cannabinoid CB1 receptor agonist, arachydonilcyclopropylamide (ACPA), in male Wistar rats was investigated. An elevated plus-maze test of anxiety was used to assess anxiety-like behaviors. The results showed that bilateral intra-amygdala injections of ACPA (0.125, 1.25 and 5 ng/rat) and the mixed dopamine D1/D2 receptor agonist, apomorphine, at different doses (0.001, 0.01 and 0.1 µg/rat) increased percentage open arm time (%OAT) and percentage open arm entries (%OAE), indicating an anxiolytic-like effect for both of the drugs. In contrast, intra-amygdala administration of the dopamine D1 receptor antagonist SCH23390 (0.5 and 1 µg/rat) and the dopamine D2 receptor antagonist, sulpiride (2 and 3 µg/rat) decreased %OAT and %OAE, suggesting an anxiogenic-like effect for both of the drugs. Interestingly, pretreatment with a sub-effective dose of apomorphine (0.0005 µg/rat) increased, while SCH23390 (0.25 µg/rat) and sulpiride (1.5 µg/rat) decreased the anxiolytic-like effect of ACPA. It can be concluded that the dopaminergic system of the amygdala may be involved, at least partly, in the anxiolytic-like effects induced by ACPA in the rat amygdala.
Collapse
Affiliation(s)
- Mohammad Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | |
Collapse
|
44
|
Abstract
Precise automatic control of food intake and energy expenditure maintains a steady weight and is fundamental to survival. The brainstem and hypothalamus are key areas within the brain that integrate peripheral signals from the gut and adipose tissue to control feeding behavior according to energy need. Gut hormones are released after a meal and signal to the brain to initiate meal termination and feelings of satiation. However, reward pathways are able to override this mechanism so that when palatable food is presented, food is consumed irrespective of energy requirements.
Collapse
Affiliation(s)
- Katherine A Simpson
- Section of Investigative Medicine, Imperial College London, Commonwealth Building, Du Cane Road, London, W12 0NN, UK
| | | |
Collapse
|
45
|
Tree K, Caravagna C, Hilaire G, Peyronnet J, Cayetanot F. Anandamide centrally depresses the respiratory rhythm generator of neonatal mice. Neuroscience 2010; 170:1098-109. [PMID: 20800658 DOI: 10.1016/j.neuroscience.2010.08.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 08/02/2010] [Accepted: 08/19/2010] [Indexed: 11/26/2022]
Abstract
Endogenous cannabinoid receptors are widely distributed throughout the CNS, including the brainstem, and modulate a variety of functions, including breathing. In adult rats, activation of cannabinoid 1 receptors has been shown to depress breathing. Here in neonatal mice, we used in vitro electrophysiology, pharmacology, and immunohistochemistry to analyse the central effects of the endocannabinoid anandamide (AEA) on the activity of the medullary respiratory rhythm generator (RRG). First of all, in vitro electrophysiology on medullary preparations has revealed that bath application of AEA (30 μM, 15 min) significantly depressed respiratory activity. Secondly, applying pre-treatments with alpha-1 (Prazosin, 5 μM, 10 min) and alpha-2 (Yohimbine, 5 μM, 10 min) adrenoceptor antagonists prior to AEA application abolished the AEA-induced depression of the RRG. Finally, immunostaining revealed a dense network of fibres positive for the cannabinoid 1 receptor in the ventrolateral medulla (VLM), a region known to contain both the RRG and the modulatory A1/C1 catecholaminergic group. Moreover, cannabinoid 1 receptor positive fibres were found in close apposition with A1/C1 catecholaminergic cells, identified by the presence of tyrosine hydroxylase. In regard of our electrophysiological, pharmacological and immunostaining results, we conclude that AEA has a central depressive effect on the neonatal RRG, probably via the medullary A1/C1 catecholaminergic neurons which are already known to modulate the respiratory rhythm generator.
Collapse
Affiliation(s)
- K Tree
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille CRN2M, Département de Physiologie Neurovégétative (PNV), Université Paul Cézanne Aix Marseille III, Faculté des Sciences et Techniques St. Jérôme, UMR, 6231 CNRS, Marseille, France
| | | | | | | | | |
Collapse
|
46
|
Physical exercise in adolescence changes CB1 cannabinoid receptor expression in the rat brain. Neurochem Int 2010; 57:492-6. [PMID: 20621146 DOI: 10.1016/j.neuint.2010.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 07/05/2010] [Indexed: 11/22/2022]
Abstract
Accumulating evidence indicates that the endocannabinoid system plays an essential role in the development and maturation of the central nervous system. Studies also have demonstrated that neural systems that regulate behavioral responses can be influenced by exercise during development. Exercise and endogenous cannabinoid activity have independently been shown to regulate brain plasticity, hence demonstrating a promising field of the endocannabinoid-exercise interaction. In order to investigate whether physical exercise during development would promote changes the brain endocannabinoid system, we investigated the cannabinoid receptor type 1 (CB1) expression in the brain of rats trained during the adolescent period. The results showed that an aerobic exercise program performed during adolescence significantly reduced the CB1 receptor expression in the striatum and hippocampal formation. These findings suggest an important link between the endocannabinoid system and physical training in adolescence.
Collapse
|
47
|
Buczynski MW, Parsons LH. Quantification of brain endocannabinoid levels: methods, interpretations and pitfalls. Br J Pharmacol 2010; 160:423-42. [PMID: 20590555 PMCID: PMC2931546 DOI: 10.1111/j.1476-5381.2010.00787.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 03/12/2010] [Accepted: 03/20/2010] [Indexed: 02/03/2023] Open
Abstract
Endocannabinoids play an important role in a diverse range of neurophysiological processes including neural development, neuroimmune function, synaptic plasticity, pain, reward and affective state. This breadth of influence and evidence for altered endocannabinoid signalling in a variety of neuropathologies has fuelled interest in the accurate quantification of these lipids in brain tissue. Established methods for endocannabinoid quantification primarily employ solvent-based lipid extraction with further sample purification by solid phase extraction. In recent years in vivo microdialysis methods have also been developed for endocannabinoid sampling from the brain interstitial space. However, considerable variability in estimates of endocannabinoid content has led to debate regarding the physiological range of concentrations present in various brain regions. This paper provides a critical review of factors that influence the quantification of brain endocannabinoid content as determined by lipid extraction from bulk tissue and by in vivo microdialysis. A variety of methodological issues are discussed including analytical approaches, endocannabinoid extraction and purification, post-mortem changes in brain endocannabinoid content, cellular reactions to microdialysis probe implantation and caveats related to lipid sampling from the extracellular space. The application of these methods for estimating brain endocannabinoid content and the effects of endocannabinoid clearance inhibition are discussed. The benefits, limitations and pitfalls associated with each approach are emphasized, with an eye toward the appropriate interpretation of data gathered by each method.
Collapse
Affiliation(s)
- Matthew W Buczynski
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
48
|
Cannabinoid-dopamine interaction in the pathophysiology and treatment of CNS disorders. CNS Neurosci Ther 2010; 16:e72-91. [PMID: 20406253 DOI: 10.1111/j.1755-5949.2010.00144.x] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Endocannabinoids and their receptors, mainly the CB(1) receptor type, function as a retrograde signaling system in many synapses within the CNS, particularly in GABAergic and glutamatergic synapses. They also play a modulatory function on dopamine (DA) transmission, although CB(1) receptors do not appear to be located in dopaminergic terminals, at least in the major brain regions receiving dopaminergic innervation, e.g., the caudate-putamen and the nucleus accumbens/prefrontal cortex. Therefore, the effects of cannabinoids on DA transmission and DA-related behaviors are generally indirect and exerted through the modulation of GABA and glutamate inputs received by dopaminergic neurons. Recent evidence suggest, however, that certain eicosanoid-derived cannabinoids may directly activate TRPV(1) receptors, which have been found in some dopaminergic pathways, thus allowing a direct regulation of DA function. Through this direct mechanism or through indirect mechanisms involving GABA or glutamate neurons, cannabinoids may interact with DA transmission in the CNS and this has an important influence in various DA-related neurobiological processes (e.g., control of movement, motivation/reward) and, particularly, on different pathologies affecting these processes like basal ganglia disorders, schizophrenia, and drug addiction. The present review will address the current literature supporting these cannabinoid-DA interactions, with emphasis in aspects dealing with the neurochemical, physiological, and pharmacological/therapeutic bases of these interactions.
Collapse
|
49
|
Yoon SS, Kim H, Choi KH, Lee BH, Lee YK, Lim SC, Choi SH, Hwang M, Kim KJ, Yang CH. Acupuncture suppresses morphine self-administration through the GABA receptors. Brain Res Bull 2010; 81:625-30. [DOI: 10.1016/j.brainresbull.2009.12.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 12/21/2009] [Indexed: 10/20/2022]
|
50
|
Kovacs K, Lajtha A, Sershen H. Effect of nicotine and cocaine on neurofilaments and receptors in whole brain tissue and synaptoneurosome preparations. Brain Res Bull 2010; 82:109-17. [DOI: 10.1016/j.brainresbull.2010.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 02/16/2010] [Accepted: 02/18/2010] [Indexed: 12/25/2022]
|