1
|
Dudek KA, Paton SEJ, Binder LB, Collignon A, Dion-Albert L, Cadoret A, Lebel M, Lavoie O, Bouchard J, Kaufmann FN, Clavet-Fournier V, Manca C, Guzmán M, Campbell M, Turecki G, Mechawar N, Flamand N, Lavoie-Cardinal F, Silvestri C, Di Marzo V, Menard C. Astrocytic cannabinoid receptor 1 promotes resilience by dampening stress-induced blood-brain barrier alterations. Nat Neurosci 2025; 28:766-782. [PMID: 40016352 PMCID: PMC11976283 DOI: 10.1038/s41593-025-01891-9] [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: 05/24/2023] [Accepted: 01/08/2025] [Indexed: 03/01/2025]
Abstract
Blood-brain barrier (BBB) alterations contribute to stress vulnerability and the development of depressive behaviors. In contrast, neurovascular adaptations underlying stress resilience remain unclear. Here we report that high expression of astrocytic cannabinoid receptor 1 (CB1) in the nucleus accumbens (NAc) shell, particularly in the end-feet ensheathing blood vessels, is associated with resilience during chronic social stress in adult male mice. Viral-mediated overexpression of Cnr1 in astrocytes of the NAc shell results in baseline anxiolytic effects and dampens stress-induced anxiety- and depression-like behaviors in male mice. It promotes the expression of vascular-related genes and reduces astrocyte inflammatory response and morphological changes following an immune challenge with the cytokine interleukin-6, linked to stress susceptibility and mood disorders. Physical exercise and antidepressant treatment increase the expression of astrocytic Cnr1 in the perivascular region in male mice. In human tissue from male donors with major depressive disorder, we observe loss of CNR1 in the NAc astrocytes. Our findings suggest a role for the astrocytic endocannabinoid system in stress responses via modulation of the BBB.
Collapse
Affiliation(s)
- Katarzyna A Dudek
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Sam E J Paton
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Luisa Bandeira Binder
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Adeline Collignon
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Laurence Dion-Albert
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Alice Cadoret
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Manon Lebel
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Olivier Lavoie
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Jonathan Bouchard
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Fernanda Neutzling Kaufmann
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Valerie Clavet-Fournier
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Claudia Manca
- Faculty of Medicine and Quebec Heart and Lung Institute, Université Laval, Quebec City, Quebec, Canada
| | - Manuel Guzmán
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, CIBERNED and IRYCIS, Madrid, Spain
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Gustavo Turecki
- Department of Psychiatry, McGill University and Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Naguib Mechawar
- Department of Psychiatry, McGill University and Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Nicolas Flamand
- Faculty of Medicine and Quebec Heart and Lung Institute, Université Laval, Quebec City, Quebec, Canada
| | - Flavie Lavoie-Cardinal
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Cristoforo Silvestri
- Faculty of Medicine and Quebec Heart and Lung Institute, Université Laval, Quebec City, Quebec, Canada
| | - Vincenzo Di Marzo
- Faculty of Medicine and Quebec Heart and Lung Institute, Université Laval, Quebec City, Quebec, Canada
- Faculty of Agricultural and Food Sciences, INAF and NUTRISS Center, Quebec City, Quebec, Canada
- Joint International Research Unit on Chemical and Biomolecular Research on the Microbiome and its Impact on Metabolic Health and Nutrition between Université Laval, Quebec City, Quebec, Canada
- Consiglio Nazionale Delle Ricerche, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Caroline Menard
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada.
| |
Collapse
|
2
|
Hebert FO, Mongeau-Pérusse V, Rizkallah E, Mahroug A, Bakouni H, Morissette F, Brissette S, Bruneau J, Dubreucq S, Jutras-Aswad D. Absence of Evidence for Sustained Effects of Daily Cannabidiol Administration on Anandamide Plasma Concentration in Individuals with Cocaine Use Disorder: Exploratory Findings from a Randomized Controlled Trial. Cannabis Cannabinoid Res 2025; 10:e341-e352. [PMID: 38770686 DOI: 10.1089/can.2023.0273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
Background: Cannabidiol (CBD) has been proposed to have a therapeutic potential over a wide range of neuropsychiatric disorders, including substance use disorders. Pre-clinical evidence suggests that CBD can increase anandamide (AEA) plasma concentration, possibly mediating some of its therapeutic properties. Whether CBD exerts such an effect on AEA in individuals with cocaine use disorder (CUD) remains unknown. Aims: To explore the sustained effects of daily CBD administration on AEA plasma concentrations compared with placebo in CUD. Methods: We used data from a randomized, double-blind, placebo-controlled trial evaluating CBD's efficacy in CUD. Seventy-eight individuals were randomized to receive a daily oral dose of 800 mg CBD (n = 40) or a placebo (n = 38). Participants stayed in an inpatient detoxification setting for 10 days, after which they were followed in an outpatient setting for 12 weeks. AEA plasma concentration was measured at baseline and at 23-h post CBD ingestion on day 8 and week 4. A generalized estimating equation model was used to assess CBD's effects on AEA, and sensitivity analyses were computed using Bayesian linear regressions. Results: Sixty-four participants were included in the analysis. Similar mean AEA plasma concentrations in both treatment groups (p = 0.357) were observed. At day 8, mean AEA plasma concentrations (± standard deviation) were 0.26 (± 0.07) ng/mL in the CBD group and 0.29 (± 0.08) ng/mL in the placebo group (p = 0.832; Bayes factor [BF] = 0.190). At week 4, they were 0.27 (± 0.09) ng/mL in the CBD group and 0.30 (± 0.09) ng/mL in the placebo group (p = 0.181; BF = 0.194). Conclusion: While not excluding any potential acute and short-term effect, daily CBD administration did not exert a sustained impact on AEA plasma concentrations in individuals with CUD compared with placebo. Registration: clinicaltrials.gov (NCT02559167).
Collapse
Affiliation(s)
| | - Violaine Mongeau-Pérusse
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Elie Rizkallah
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Amani Mahroug
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
| | - Hamzah Bakouni
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Florence Morissette
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Suzanne Brissette
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Family and Emergency Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Julie Bruneau
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Family and Emergency Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Simon Dubreucq
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Didier Jutras-Aswad
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| |
Collapse
|
3
|
Serra V, Aroni S, Bortolato M, Frau R, Melis M. Endocannabinoid-dependent decrease of GABAergic transmission on dopaminergic neurons is associated with susceptibility to cocaine stimulant effects in pre-adolescent male MAOA hypomorphic mice exposed to early life stress. Neuropharmacology 2023; 233:109548. [PMID: 37080337 DOI: 10.1016/j.neuropharm.2023.109548] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/04/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Vulnerability to cocaine use disorder depends upon a combination of genetic and environmental risk factors. While early life adversity is a critical environmental vulnerability factor for drug misuse, allelic variants of the monoamine oxidase A (MAOA) gene have been shown to moderate its influence on the risk of drug-related problems. However, data on the interactions between MAOA variants and early life stress (ES) with respect to predisposition to cocaine abuse are limited. Here, we show that a mouse model capturing the interaction of genetic (low-activity alleles of the Maoa gene; MAOANeo) and environmental (i.e., ES) vulnerability factors displays an increased sensitivity to repeated in vivo cocaine psychomotor stimulant actions associated with a reduction of GABAA receptor-mediated inhibition of dopamine neurons of the ventral tegmental area (VTA). Depolarization-induced suppression of inhibition (DSI), a 2-arachidonoylglycerol (2AG)-dependent form of short-term plasticity, also becomes readily expressed by dopamine neurons from male MAOANeo ES mice repeatedly treated with cocaine. The activation of either dopamine D2 or CB1 receptors contributes to cocaine-induced DSI expression, decreased GABA synaptic efficacy, and hyperlocomotion. Next, in vivo pharmacological enhancement of 2AG signaling during repeated cocaine exposure occludes its actions both in vivo and ex vivo. This data extends our knowledge of the multifaceted sequelae imposed by this gene-environment interaction in VTA dopamine neurons of male pre-adolescent mice and contributes to our understanding of neural mechanisms of vulnerability for early onset cocaine use.
Collapse
Affiliation(s)
- Valeria Serra
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy
| | - Sonia Aroni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, 84112, USA
| | - Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy.
| |
Collapse
|
4
|
García-Cárceles J, Vázquez-Villa H, Brea J, Ladron de Guevara-Miranda D, Cincilla G, Sánchez-Martínez M, Sánchez-Merino A, Algar S, Teresa de Los Frailes M, Roberts RS, Ballesteros JA, Rodríguez de Fonseca F, Benhamú B, Loza MI, López-Rodríguez ML. 2-(Fluoromethoxy)-4'-( S-methanesulfonimidoyl)-1,1'-biphenyl (UCM-1306), an Orally Bioavailable Positive Allosteric Modulator of the Human Dopamine D 1 Receptor for Parkinson's Disease. J Med Chem 2022; 65:12256-12272. [PMID: 36044544 PMCID: PMC9511493 DOI: 10.1021/acs.jmedchem.2c00949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
![]()
Tolerance development caused by dopamine replacement
with l-DOPA and therapeutic drawbacks upon activation of
dopaminergic receptors
with orthosteric agonists reveal a significant unmet need for safe
and effective treatment of Parkinson’s disease. In search for
selective modulators of the D1 receptor, the screening
of a chemical library and subsequent medicinal chemistry program around
an identified hit resulted in new synthetic compound 26 [UCM-1306, 2-(fluoromethoxy)-4′-(S-methanesulfonimidoyl)-1,1′-biphenyl]
that increases the dopamine maximal effect in a dose-dependent manner
in human and mouse D1 receptors, is inactive in the absence
of dopamine, modulates dopamine affinity for the receptor, exhibits
subtype selectivity, and displays low binding competition with orthosteric
ligands. The new allosteric modulator potentiates cocaine-induced
locomotion and enhances l-DOPA recovery of decreased locomotor
activity in reserpinized mice after oral administration. The behavior
of compound 26 supports the interest of a positive allosteric
modulator of the D1 receptor as a promising therapeutic
approach for Parkinson’s disease.
Collapse
Affiliation(s)
- Javier García-Cárceles
- Departamento de Química Orgánica, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Henar Vázquez-Villa
- Departamento de Química Orgánica, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - José Brea
- Biofarma Research Group, USEF Screening Platform, CIMUS, USC, E-15782 Santiago de Compostela, Spain
| | | | - Giovanni Cincilla
- Molomics S.L., Parc Científic de Barcelona, Baldiri Reixac 4-8, E-08028 Barcelona, Spain
| | | | - Anabel Sánchez-Merino
- Departamento de Química Orgánica, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Sergio Algar
- Departamento de Química Orgánica, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - María Teresa de Los Frailes
- Fundación Kærtor, Edificio EMPRENDIA, Planta 2, Oficina 4. Campus Vida, E-15706 Santiago de Compostela, Spain
| | - Richard S Roberts
- Fundación Kærtor, Edificio EMPRENDIA, Planta 2, Oficina 4. Campus Vida, E-15706 Santiago de Compostela, Spain
| | | | | | - Bellinda Benhamú
- Departamento de Química Orgánica, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - María I Loza
- Biofarma Research Group, USEF Screening Platform, CIMUS, USC, E-15782 Santiago de Compostela, Spain.,Fundación Kærtor, Edificio EMPRENDIA, Planta 2, Oficina 4. Campus Vida, E-15706 Santiago de Compostela, Spain
| | | |
Collapse
|
5
|
Navarrete F, García-Gutiérrez MS, Gasparyan A, Navarro D, López-Picón F, Morcuende Á, Femenía T, Manzanares J. Biomarkers of the Endocannabinoid System in Substance Use Disorders. Biomolecules 2022; 12:biom12030396. [PMID: 35327588 PMCID: PMC8946268 DOI: 10.3390/biom12030396] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/21/2022] [Accepted: 02/28/2022] [Indexed: 02/04/2023] Open
Abstract
Despite substance use disorders (SUD) being one of the leading causes of disability and mortality globally, available therapeutic approaches remain ineffective. The difficulty in accurately characterizing the neurobiological mechanisms involved with a purely qualitative diagnosis is an obstacle to improving the classification and treatment of SUD. In this regard, identifying central and peripheral biomarkers is essential to diagnosing the severity of drug dependence, monitoring therapeutic efficacy, predicting treatment response, and enhancing the development of safer and more effective pharmacological tools. In recent years, the crucial role that the endocannabinoid system (ECS) plays in regulating the reinforcing and motivational properties of drugs of abuse has been described. This has led to studies characterizing ECS alterations after exposure to various substances to identify biomarkers with potential diagnostic, prognostic, or therapeutic utility. This review aims to compile the primary evidence available from rodent and clinical studies on how the ECS components are modified in the context of different substance-related disorders, gathering data from genetic, molecular, functional, and neuroimaging experimental approaches. Finally, this report concludes that additional translational research is needed to further characterize the modifications of the ECS in the context of SUD, and their potential usefulness in the necessary search for biomarkers.
Collapse
Affiliation(s)
- Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (A.G.); (D.N.); (Á.M.); (T.F.)
- Departamento de Medicina Clínica, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Universidad Miguel Hernández, 03010 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - María S. García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (A.G.); (D.N.); (Á.M.); (T.F.)
- Departamento de Medicina Clínica, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Universidad Miguel Hernández, 03010 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (A.G.); (D.N.); (Á.M.); (T.F.)
- Departamento de Medicina Clínica, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Universidad Miguel Hernández, 03010 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Daniela Navarro
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (A.G.); (D.N.); (Á.M.); (T.F.)
- Departamento de Medicina Clínica, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Universidad Miguel Hernández, 03010 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Francisco López-Picón
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, 20520 Turku, Finland;
| | - Álvaro Morcuende
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (A.G.); (D.N.); (Á.M.); (T.F.)
| | - Teresa Femenía
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (A.G.); (D.N.); (Á.M.); (T.F.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (A.G.); (D.N.); (Á.M.); (T.F.)
- Departamento de Medicina Clínica, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Universidad Miguel Hernández, 03010 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-965-919-248
| |
Collapse
|
6
|
De Sa Nogueira D, Bourdy R, Alcala-Vida R, Filliol D, Andry V, Goumon Y, Zwiller J, Romieu P, Merienne K, Olmstead MC, Befort K. Hippocampal Cannabinoid 1 Receptors Are Modulated Following Cocaine Self-administration in Male Rats. Mol Neurobiol 2022; 59:1896-1911. [PMID: 35032317 DOI: 10.1007/s12035-022-02722-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023]
Abstract
Cocaine addiction is a complex pathology inducing long-term neuroplastic changes that, in turn, contribute to maladaptive behaviors. This behavioral dysregulation is associated with transcriptional reprogramming in brain reward circuitry, although the mechanisms underlying this modulation remain poorly understood. The endogenous cannabinoid system may play a role in this process in that cannabinoid mechanisms modulate drug reward and contribute to cocaine-induced neural adaptations. In this study, we investigated whether cocaine self-administration induces long-term adaptations, including transcriptional modifications and associated epigenetic processes. We first examined endocannabinoid gene expression in reward-related brain regions of the rat following self-administered (0.33 mg/kg intravenous, FR1, 10 days) cocaine injections. Interestingly, we found increased Cnr1 expression in several structures, including prefrontal cortex, nucleus accumbens, dorsal striatum, hippocampus, habenula, amygdala, lateral hypothalamus, ventral tegmental area, and rostromedial tegmental nucleus, with most pronounced effects in the hippocampus. Endocannabinoid levels, measured by mass spectrometry, were also altered in this structure. Chromatin immunoprecipitation followed by qPCR in the hippocampus revealed that two activating histone marks, H3K4Me3 and H3K27Ac, were enriched at specific endocannabinoid genes following cocaine intake. Targeting CB1 receptors using chromosome conformation capture, we highlighted spatial chromatin re-organization in the hippocampus, as well as in the nucleus accumbens, suggesting that destabilization of the chromatin may contribute to neuronal responses to cocaine. Overall, our results highlight a key role for the hippocampus in cocaine-induced plasticity and broaden the understanding of neuronal alterations associated with endocannabinoid signaling. The latter suggests that epigenetic modifications contribute to maladaptive behaviors associated with chronic drug use.
Collapse
Affiliation(s)
- David De Sa Nogueira
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France.,Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Romain Bourdy
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Rafael Alcala-Vida
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Dominique Filliol
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 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
| | - Jean Zwiller
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Pascal Romieu
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Karine Merienne
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Mary C Olmstead
- Department of Psychology, Center for Neuroscience Studies, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Katia Befort
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France.
| |
Collapse
|
7
|
Gish A, Wiart JF, Turpin E, Allorge D, Gaulier JM. État de l’art et intérêt des dosages plasmatiques des substances endocannabinoïdes et endocannabinoïdes-like. TOXICOLOGIE ANALYTIQUE ET CLINIQUE 2021. [DOI: 10.1016/j.toxac.2021.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
8
|
Mitra S, Gobira PH, Werner CT, Martin JA, Iida M, Thomas SA, Erias K, Miracle S, Lafargue C, An C, Dietz DM. A role for the endocannabinoid enzymes monoacylglycerol and diacylglycerol lipases in cue-induced cocaine craving following prolonged abstinence. Addict Biol 2021; 26:e13007. [PMID: 33496035 PMCID: PMC11000690 DOI: 10.1111/adb.13007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 12/25/2020] [Accepted: 01/12/2021] [Indexed: 01/01/2023]
Abstract
Following exposure to drugs of abuse, long-term neuroadaptations underlie persistent risk to relapse. Endocannabinoid signaling has been associated with drug-induced neuroadaptations, but the role of lipases that mediate endocannabinoid biosynthesis and metabolism in regulating relapse behaviors following prolonged periods of drug abstinence has not been examined. Here, we investigated how pharmacological manipulation of lipases involved in regulating the expression of the endocannabinoid 2-AG in the nucleus accumbens (NAc) influence cocaine relapse via discrete neuroadaptations. At prolonged abstinence (30 days) from cocaine self-administration, there is an increase in the NAc levels of diacylglycerol lipase (DAGL), the enzyme responsible for the synthesis of the endocannabinoid 2-AG, along with decreased levels of monoacylglycerol lipase (MAGL), which hydrolyzes 2-AG. Since endocannabinoid-mediated behavioral plasticity involves phosphatase dysregulation, we examined the phosphatase calcineurin after 30 days of abstinence and found decreased expression in the NAc, which we demonstrate is regulated through the transcription factor EGR1. Intra-NAc pharmacological manipulation of DAGL and MAGL with inhibitors DO-34 and URB-602, respectively, bidirectionally regulated cue-induced cocaine seeking and altered the phosphostatus of translational initiation factor, eIF2α. Finally, we found that cocaine seeking 30 days after abstinence leads to decreased phosphorylation of eIF2α and reduced expression of its downstream target NPAS4, a protein involved in experience-dependent neuronal plasticity. Together, our findings demonstrate that lipases that regulate 2-AG expression influence transcriptional and translational changes in the NAc related to drug relapse vulnerability.
Collapse
Affiliation(s)
- Swarup Mitra
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
- These authors contributed equally to this work
| | - Pedro H. Gobira
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- These authors contributed equally to this work
| | - Craig T. Werner
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Jennifer A. Martin
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Madoka Iida
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Shruthi A. Thomas
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Kyra Erias
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Sophia Miracle
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Charles Lafargue
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Chunna An
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
| | - David M. Dietz
- Department of Pharmacology and Toxicology, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
- Department of Psychology, The State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
9
|
Miquel M, Gil-Miravet I, Guarque-Chabrera J. The Cerebellum on Cocaine. Front Syst Neurosci 2020; 14:586574. [PMID: 33192350 PMCID: PMC7641605 DOI: 10.3389/fnsys.2020.586574] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/29/2020] [Indexed: 12/30/2022] Open
Abstract
The traditional cerebellum’s role has been linked to the high computational demands for sensorimotor control. However, several findings have pointed to its involvement in executive and emotional functions in the last decades. First in 2009 and then, in 2016, we raised why we should consider the cerebellum when thinking about drug addiction. A decade later, mounting evidence strongly suggests the cerebellar involvement in this disorder. Nevertheless, direct evidence is still partial and related mainly to drug-induced reward memory, but recent results about cerebellar functions may provide new insights into its role in addiction. The present review does not intend to be a compelling revision on available findings, as we did in the two previous reviews. This minireview focuses on specific findings of the cerebellum’s role in drug-related reward memories and the way ahead for future research. The results discussed here provide grounds for involving the cerebellar cortex’s apical region in regulating behavior driven by drug-cue associations. They also suggest that the cerebellar cortex dysfunction may facilitate drug-induced learning by increasing glutamatergic output from the deep cerebellar nucleus (DCN) to the ventral tegmental area (VTA) and neural activity in its projecting areas.
Collapse
Affiliation(s)
- Marta Miquel
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Spain
| | - Isis Gil-Miravet
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Spain
| | | |
Collapse
|
10
|
Altamura AC, Delvecchio G, Marotta G, Oldani L, Pigoni A, Ciappolino V, Caletti E, Rovera C, Dobrea C, Arici C, Benatti B, Camuri G, Prunas C, Paoli RA, Dell'osso B, Cinnante C, Triulzi FM, Brambilla P. Structural and metabolic differentiation between bipolar disorder with psychosis and substance-induced psychosis: An integrated MRI/PET study. Eur Psychiatry 2016; 41:85-94. [PMID: 28049086 DOI: 10.1016/j.eurpsy.2016.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/20/2016] [Accepted: 09/24/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Bipolar disorder (BD) may be characterized by the presence of psychotic symptoms and comorbid substance abuse. In this context, structural and metabolic dysfunctions have been reported in both BD with psychosis and addiction, separately. In this study, we aimed at identifying neural substrates differentiating psychotic BD, with or without substance abuse, versus substance-induced psychosis (SIP) by coupling, for the first time, magnetic resonance imaging (MRI) and positron emission tomography (PET). METHODS Twenty-seven BD type I psychotic patients with (n=10) or without (n=17) substance abuse, 16 SIP patients and 54 healthy controls were enrolled in this study. 3T MRI and 18-FDG-PET scanning were acquired. RESULTS Gray matter (GM) volume and cerebral metabolism reductions in temporal cortices were observed in all patients compared to healthy controls. Moreover, a distinct pattern of fronto-limbic alterations were found in patients with substance abuse. Specifically, BD patients with substance abuse showed volume reductions in ventrolateral prefrontal cortex, anterior cingulate, insula and thalamus, whereas SIP patients in dorsolateral prefrontal cortex and posterior cingulate. Common alterations in cerebellum, parahippocampus and posterior cingulate were found in both BD with substance abuse and SIP. Finally, a unique pattern of GM volumes reduction, with concomitant increased of striatal metabolism, were observed in SIP patients. CONCLUSIONS These findings contribute to shed light on the identification of common and distinct neural markers associated with bipolar psychosis and substance abuse. Future longitudinal studies should explore the effect of single substances of abuse in patients at the first-episode of BD and substance-induced psychosis.
Collapse
Affiliation(s)
- A C Altamura
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - G Delvecchio
- Scientific Institute IRCCS "E. Medea", San Vito al Tagliamento (PN), Italy
| | - G Marotta
- Department of Services, Neuroradiology Unit, Nuclear Medicine Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - L Oldani
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - A Pigoni
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - V Ciappolino
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - E Caletti
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - C Rovera
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - C Dobrea
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - C Arici
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - B Benatti
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - G Camuri
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - C Prunas
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - R A Paoli
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - B Dell'osso
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy; Department of Psychiatry, Bipolar Disorders Clinic, Stanford University, CA, USA
| | - C Cinnante
- Department of Services, Neuroradiology Unit, Nuclear Medicine Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - F M Triulzi
- Department of Services, Neuroradiology Unit, Nuclear Medicine Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - P Brambilla
- Department of Neurosciences and Mental Health, Institute of Psychiatry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy; Department of Psychiatry and Behavioural Neurosciences, University of Texas at Houston, Houston, TX, USA.
| |
Collapse
|
11
|
Márquez J, Campos-Sandoval JA, Peñalver A, Matés JM, Segura JA, Blanco E, Alonso FJ, de Fonseca FR. Glutamate and Brain Glutaminases in Drug Addiction. Neurochem Res 2016; 42:846-857. [DOI: 10.1007/s11064-016-2137-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/12/2016] [Accepted: 12/08/2016] [Indexed: 10/20/2022]
|
12
|
Have we been ignoring the elephant in the room? Seven arguments for considering the cerebellum as part of addiction circuitry. Neurosci Biobehav Rev 2015; 60:1-11. [PMID: 26602022 DOI: 10.1016/j.neubiorev.2015.11.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 11/09/2015] [Accepted: 11/16/2015] [Indexed: 12/23/2022]
Abstract
Addiction involves alterations in multiple brain regions that are associated with functions such as memory, motivation and executive control. Indeed, it is now well accepted that addictive drugs produce long-lasting molecular and structural plasticity changes in corticostriatal-limbic loops. However, there are brain regions that might be relevant to addiction other than the prefrontal cortex, amygdala, hippocampus and basal ganglia. In addition to these circuits, a growing amount of data suggests the involvement of the cerebellum in many of the brain functions affected in addicts, though this region has been overlooked, traditionally, in the addiction field. Therefore, in the present review we provide seven arguments as to why we should consider the cerebellum in drug addiction. We present and discuss compelling evidence about the effects of drugs of abuse on cerebellar plasticity, the involvement of the cerebellum in drug-induced cue-related memories, and several findings showing that the instrumental memory and executive functions also recruit the cerebellar circuitry. In addition, a hypothetical model of the cerebellum's role relative to other areas within corticostriatal-limbic networks is also provided. Our goal is not to review animal and human studies exhaustively but to support the inclusion of cerebellar alterations as a part of the physiopathology of addiction disorder.
Collapse
|
13
|
Abstract
Brain endocannabinoid (eCB) signalling influences the motivation for natural rewards (such as palatable food, sexual activity and social interaction) and modulates the rewarding effects of addictive drugs. Pathological forms of natural and drug-induced reward are associated with dysregulated eCB signalling that may derive from pre-existing genetic factors or from prolonged drug exposure. Impaired eCB signalling contributes to dysregulated synaptic plasticity, increased stress responsivity, negative emotional states and cravings that propel addiction. Understanding the contributions of eCB disruptions to behavioural and physiological traits provides insight into the eCB influence on addiction vulnerability.
Collapse
Affiliation(s)
- Loren H Parsons
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Yasmin L Hurd
- Friedman Brain Institute, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York City, New York 10029, USA
| |
Collapse
|
14
|
López-Pedrajas R, Ramírez-Lamelas DT, Muriach B, Sánchez-Villarejo MV, Almansa I, Vidal-Gil L, Romero FJ, Barcia JM, Muriach M. Cocaine promotes oxidative stress and microglial-macrophage activation in rat cerebellum. Front Cell Neurosci 2015; 9:279. [PMID: 26283916 PMCID: PMC4516895 DOI: 10.3389/fncel.2015.00279] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/09/2015] [Indexed: 12/14/2022] Open
Abstract
Different mechanisms have been suggested for cocaine neurotoxicity, including oxidative stress alterations. Nuclear factor kappa B (NF-κB), considered a sensor of oxidative stress and inflammation, is involved in drug toxicity and addiction. NF-κB is a key mediator for immune responses that induces microglial/macrophage activation under inflammatory processes and neuronal injury/degeneration. Although cerebellum is commonly associated to motor control, muscular tone, and balance. Its relation with addiction is getting relevance, being associated to compulsive and perseverative behaviors. Some reports indicate that cerebellar microglial activation induced by cannabis or ethanol, promote cerebellar alterations and these alterations could be associated to addictive-related behaviors. After considering the effects of some drugs on cerebellum, the aim of the present work analyzes pro-inflammatory changes after cocaine exposure. Rats received daily 15 mg/kg cocaine i.p., for 18 days. Reduced and oxidized forms of glutathione (GSH) and oxidized glutathione (GSSG), glutathione peroxidase (GPx) activity and glutamate were determined in cerebellar homogenates. NF-κB activity, CD68, and GFAP expression were determined. Cerebellar GPx activity and GSH/GSSG ratio are significantly decreased after cocaine exposure. A significant increase of glutamate concentration is also observed. Interestingly, increased NF-κB activity is also accompanied by an increased expression of the lysosomal mononuclear phagocytic marker ED1 without GFAP alterations. Current trends in addiction biology are focusing on the role of cerebellum on addictive behaviors. Cocaine-induced cerebellar changes described herein fit with previosus data showing cerebellar alterations on addict subjects and support the proposed role of cerebelum in addiction.
Collapse
Affiliation(s)
- Rosa López-Pedrajas
- Instituto de Ciencias Biomédicas, Departamento de Ciencias Biomédicas, Universidad CEU Cardenal Herrera Moncada, Valencia, Spain
| | - Dolores T Ramírez-Lamelas
- Instituto de Ciencias Biomédicas, Departamento de Ciencias Biomédicas, Universidad CEU Cardenal Herrera Moncada, Valencia, Spain
| | - Borja Muriach
- Instituto de Ciencias Biomédicas, Departamento de Ciencias Biomédicas, Universidad CEU Cardenal Herrera Moncada, Valencia, Spain
| | - María V Sánchez-Villarejo
- Instituto de Ciencias Biomédicas, Departamento de Ciencias Biomédicas, Universidad CEU Cardenal Herrera Moncada, Valencia, Spain
| | - Inmaculada Almansa
- Instituto de Ciencias Biomédicas, Departamento de Ciencias Biomédicas, Universidad CEU Cardenal Herrera Moncada, Valencia, Spain
| | - Lorena Vidal-Gil
- Structure and Function of the Human Body, Facultad de Medicina y Odontología, Universidad Católica de Valencia 'San Vicente Mártir,' Valencia Spain
| | - Francisco J Romero
- Structure and Function of the Human Body, Facultad de Medicina y Odontología, Universidad Católica de Valencia 'San Vicente Mártir,' Valencia Spain
| | - Jorge M Barcia
- Structure and Function of the Human Body, Facultad de Medicina y Odontología, Universidad Católica de Valencia 'San Vicente Mártir,' Valencia Spain
| | - María Muriach
- UP Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Castellón Spain
| |
Collapse
|
15
|
Marinho EAV, Oliveira-Lima AJ, Santos R, Hollais AW, Baldaia MA, Wuo-Silva R, Yokoyama TS, Takatsu-Coleman AL, Patti CL, Longo BM, Berro LF, Frussa-Filho R. Effects of rimonabant on the development of single dose-induced behavioral sensitization to ethanol, morphine and cocaine in mice. Prog Neuropsychopharmacol Biol Psychiatry 2015; 58:22-31. [PMID: 25496830 DOI: 10.1016/j.pnpbp.2014.11.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/01/2014] [Accepted: 11/18/2014] [Indexed: 11/19/2022]
Abstract
RATIONALE The endocannabinoid system has been implicated in the neurobiological mechanism underlying drug addiction, especially the primary rewarding dopamine-dependent processes. Therefore, endocannabinoid receptor antagonists, such as the CB1 cannabinoid antagonist rimonabant, have been proposed as candidates for preventive addiction therapies. OBJECTIVES Investigate the possible involvement of CB1 receptors in the development of behavioral sensitization to ethanol, morphine and cocaine in mice. METHODS We compared the effects of different doses of rimonabant (0.3, 1, 3 and 10mg/kg) on spontaneous locomotor activity in the open-field, hyperlocomotion induced by acute administration of ethanol (1.8g/kg), morphine (20mg/kg) or cocaine (10mg/kg) and on subsequent drug-induced locomotor sensitization using a two-injection protocol in mice. We also investigated a possible depressive-like effect of an acute rimonabant challenge at the highest dose and its potential anxiogenic property. RESULTS At the highest dose, rimonabant abolished ethanol- and cocaine-induced hyperlocomotion and behavioral sensitization without modifying spontaneous and central locomotor activity or inducing depressive-like behavior on the forced swim test in mice. The other doses of rimonabant also selectively blocked acute ethanol-induced central hyperlocomotion. Although rimonabant at 0.3 and 1mg/kg potentiated the central hyperlocomotion induced by acute morphine injection, it was effective in attenuating morphine-induced behavioral sensitization at all doses. CONCLUSIONS Because the neural basis of behavioral sensitization has been proposed to correspond to some components of addiction, our findings indicate that the endocannabinoid system might be involved in ethanol, cocaine and morphine abuse.
Collapse
Affiliation(s)
- Eduardo A V Marinho
- Departamento de Ciências da Saúde, Universidade Estadual de Santa Cruz - UESC, Ilhéus, BA, Brazil.
| | | | - Renan Santos
- Departamento de Fisiologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - André W Hollais
- Departamento de Fisiologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Marilia A Baldaia
- Departamento de Farmacologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Raphael Wuo-Silva
- Departamento de Fisiologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Thais S Yokoyama
- Departamento de Fisiologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - André L Takatsu-Coleman
- Departamento de Psicobiologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Camilla L Patti
- Departamento de Farmacologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Beatriz M Longo
- Departamento de Fisiologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil; Departamento de Farmacologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Laís F Berro
- Departamento de Psicobiologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil.
| | - Roberto Frussa-Filho
- Departamento de Farmacologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil; Departamento de Psicobiologia, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| |
Collapse
|
16
|
Herman MA, Roberto M. The addicted brain: understanding the neurophysiological mechanisms of addictive disorders. Front Integr Neurosci 2015; 9:18. [PMID: 25852502 PMCID: PMC4365688 DOI: 10.3389/fnint.2015.00018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 02/20/2015] [Indexed: 01/29/2023] Open
Affiliation(s)
- Melissa A Herman
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| |
Collapse
|