1
|
Peeters LD, Wills LJ, Cuozzo AM, Ivanich KL, Turney SE, Bullock LP, Price RM, Gass JT, Brown RW. Modulation of mGlu5 reduces rewarding associative properties of nicotine via changes in mesolimbic plasticity: Relevance to comorbid cigarette smoking in psychosis. Pharmacol Biochem Behav 2024; 239:173752. [PMID: 38521210 PMCID: PMC11088493 DOI: 10.1016/j.pbb.2024.173752] [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: 12/01/2023] [Revised: 02/13/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
RATIONALE Antipsychotic medications that are used to treat psychosis are often limited in their efficacy by high rates of severe side effects. Treatment success in schizophrenia is further complicated by high rates of comorbid nicotine use. Dopamine D2 heteroreceptor complexes have recently emerged as targets for the development of more efficacious pharmaceutical treatments for schizophrenia. OBJECTIVE The current study sought to explore the use of the positive allosteric modulator of the mGlu5 receptor 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) as a treatment to reduce symptoms related to psychosis and comorbid nicotine use. METHODS Neonatal treatment of animals with the dopamine D2-like receptor agonist quinpirole (NQ) from postnatal day (P)1-21 produces a lifelong increase in D2 receptor sensitivity, showing relevance to psychosis and comorbid tobacco use disorder. Following an 8-day conditioning paradigm, brain tissue in the mesolimbic pathway was analyzed for several plasticity markers, including brain derived neurotrophic factor (BDNF), phosphorylated p70 ribosomal S6 kinase (phospho-p70S6K), and cadherin-13 (Cdh13). RESULTS Pretreatment with CDPPB was effective to block enhanced nicotine conditioned place preference observed in NQ-treated animals. Pretreatment was additionally effective to block the nicotine-induced increase in BDNF and sex-dependent increases in cadherin-13 in the ventral tegmental area (VTA), as well as increased phospho-p70S6K in the nucleus accumbens (NAcc) shell found in NQ-treated animals. CONCLUSION In conjunction with prior work, the current study suggests positive allosteric modulation of the mGlu5 receptor, an emerging target for schizophrenia therapeutics, may be effective for the treatment of comorbid nicotine abuse in psychosis.
Collapse
Affiliation(s)
- Loren D Peeters
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Liza J Wills
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Anthony M Cuozzo
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Kira L Ivanich
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Seth E Turney
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Luke P Bullock
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Robert M Price
- Department of Mathematics and Statistics, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Justin T Gass
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Russell W Brown
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America.
| |
Collapse
|
2
|
Mohammad Aghaei A, Saali A, Canas MA, Weleff J, D'Souza DC, Angarita GA, Bassir Nia A. Dysregulation of the endogenous cannabinoid system following opioid exposure. Psychiatry Res 2023; 330:115586. [PMID: 37931479 PMCID: PMC10842415 DOI: 10.1016/j.psychres.2023.115586] [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: 07/15/2023] [Revised: 10/05/2023] [Accepted: 10/28/2023] [Indexed: 11/08/2023]
Abstract
Rates of opioid-related deaths and overdoses in the United States are at record-high levels. Thus, novel neurobiological targets for the treatment of OUD are greatly needed. Given the close interaction between the endogenous opioid system and the endocannabinoid system (ECS), targeting the ECS may have therapeutic potential in OUD. The various components of the ECS, including cannabinoid receptors, their lipid-derived endogenous ligands (endocannabinoids [eCBs]), and the related enzymes, present potential targets for developing new medications in OUD treatment. The purpose of this paper is to review the clinical and preclinical literature on the dysregulation of the ECS after exposure to opioids. We review the evidence of ECS dysregulation across various study types, exposure protocols, and measurement protocols and summarize the evidence for dysregulation of ECS components at specific brain regions. Preclinical research has shown that opioids disrupt various ECS components that are region-specific. However, the results in the literature are highly heterogenous and sometimes contradictory, possibly due to variety of different methods used. Further research is needed before a confident conclusion could be made on how exposure to opioids can affect ECS components in various brain regions.
Collapse
Affiliation(s)
- Ardavan Mohammad Aghaei
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, United States
| | - Alexandra Saali
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | | | - Jeremy Weleff
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, United States
| | - Deepak Cyril D'Souza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, United States; VA Connecticut Healthcare System, West Haven, CT, United States
| | - Gustavo A Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, United States
| | - Anahita Bassir Nia
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, United States.
| |
Collapse
|
3
|
Davies RA, Barbee BR, Garcia-Sifuentes Y, Butkovich LM, Gourley SL. Subunit-selective PI3-kinase control of action strategies in the medial prefrontal cortex. Neurobiol Learn Mem 2023; 203:107789. [PMID: 37328026 PMCID: PMC10527156 DOI: 10.1016/j.nlm.2023.107789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/07/2023] [Accepted: 06/11/2023] [Indexed: 06/18/2023]
Abstract
PI3-kinase (PI3K) is an intracellular signaling complex that is stimulated upon cocaine exposure and linked with the behavioral consequences of cocaine. We recently genetically silenced the PI3K p110β subunit in the medial prefrontal cortex following repeated cocaine in mice, reinstating the capacity of these mice to engage in prospective goal-seeking behavior. In the present short report, we address two follow-up hypotheses: 1) The control of decision-making behavior by PI3K p110β is attributable to neuronal signaling, and 2) PI3K p110β in the healthy (i.e., drug-naïve) medial prefrontal cortex has functional consequences in the control of reward-related decision-making strategies. In Experiment 1, we found that silencing neuronal p110β improved action flexibility following cocaine. In Experiment 2, we reduced PI3K p110β in drug-naïve mice that were extensively trained to respond for food reinforcers. Gene silencing caused mice to abandon goal-seeking strategies, unmasking habit-based behaviors that were propelled by interactions with the nucleus accumbens. Thus, PI3K control of goal-directed action strategies appears to act in accordance with an inverted U-shaped function, with "too much" (following cocaine) or "too little" (following p110β subunit silencing) obstructing goal seeking and causing mice to defer to habit-like response sequences.
Collapse
Affiliation(s)
- Rachel A Davies
- Department of Pediatrics, Emory University School of Medicine, Emory National Primate Research Center, Emory University, Children's Healthcare of Atlanta, USA
| | - Britton R Barbee
- Department of Pediatrics, Emory University School of Medicine, Emory National Primate Research Center, Emory University, Children's Healthcare of Atlanta, USA; Graduate Program in Molecular and Systems Pharmacology, Emory University, USA
| | - Yesenia Garcia-Sifuentes
- Department of Pediatrics, Emory University School of Medicine, Emory National Primate Research Center, Emory University, Children's Healthcare of Atlanta, USA; Graduate Program in Neuroscience, Emory University, USA
| | - Laura M Butkovich
- Department of Pediatrics, Emory University School of Medicine, Emory National Primate Research Center, Emory University, Children's Healthcare of Atlanta, USA
| | - Shannon L Gourley
- Department of Pediatrics, Emory University School of Medicine, Emory National Primate Research Center, Emory University, Children's Healthcare of Atlanta, USA; Graduate Program in Molecular and Systems Pharmacology, Emory University, USA; Graduate Program in Neuroscience, Emory University, USA.
| |
Collapse
|
4
|
Mendez EF, Grimm SL, Stertz L, Gorski D, Movva SV, Najera K, Moriel K, Meyer TD, Fries GR, Coarfa C, Walss-Bass C. A human stem cell-derived neuronal model of morphine exposure reflects brain dysregulation in opioid use disorder: Transcriptomic and epigenetic characterization of postmortem-derived iPSC neurons. Front Psychiatry 2023; 14:1070556. [PMID: 36873219 PMCID: PMC9978009 DOI: 10.3389/fpsyt.2023.1070556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/18/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Human-derived induced pluripotent stem cell (iPSC) models of brain promise to advance our understanding of neurotoxic consequences of drug use. However, how well these models recapitulate the actual genomic landscape and cell function, as well as the drug-induced alterations, remains to be established. New in vitro models of drug exposure are needed to advance our understanding of how to protect or reverse molecular changes related to substance use disorders. Methods We engineered a novel induced pluripotent stem cell-derived model of neural progenitor cells and neurons from cultured postmortem human skin fibroblasts, and directly compared these to isogenic brain tissue from the donor source. We assessed the maturity of the cell models across differentiation from stem cells to neurons using RNA cell type and maturity deconvolution analyses as well as DNA methylation epigenetic clocks trained on adult and fetal human tissue. As proof-of-concept of this model's utility for substance use disorder studies, we compared morphine- and cocaine-treated neurons to gene expression signatures in postmortem Opioid Use Disorder (OUD) and Cocaine Use Disorder (CUD) brains, respectively. Results Within each human subject (N = 2, 2 clones each), brain frontal cortex epigenetic age parallels that of skin fibroblasts and closely approximates the donor's chronological age; stem cell induction from fibroblast cells effectively sets the epigenetic clock to an embryonic age; and differentiation of stem cells to neural progenitor cells and then to neurons progressively matures the cells via DNA methylation and RNA gene expression readouts. In neurons derived from an individual who died of opioid overdose, morphine treatment induced alterations in gene expression similar to those previously observed in OUD ex-vivo brain tissue, including differential expression of the immediate early gene EGR1, which is known to be dysregulated by opioid use. Discussion In summary, we introduce an iPSC model generated from human postmortem fibroblasts that can be directly compared to corresponding isogenic brain tissue and can be used to model perturbagen exposure such as that seen in opioid use disorder. Future studies with this and other postmortem-derived brain cellular models, including cerebral organoids, can be an invaluable tool for understanding mechanisms of drug-induced brain alterations.
Collapse
Affiliation(s)
- Emily F. Mendez
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sandra L. Grimm
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, United States
| | - Laura Stertz
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Damian Gorski
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas Medical School at Houston, Houston, TX, United States
| | - Sai V. Movva
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Katherine Najera
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Karla Moriel
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Thomas D. Meyer
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Gabriel R. Fries
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Cristian Coarfa
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, United States
| | - Consuelo Walss-Bass
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
5
|
Scheyer A, Yasmin F, Naskar S, Patel S. Endocannabinoids at the synapse and beyond: implications for neuropsychiatric disease pathophysiology and treatment. Neuropsychopharmacology 2023; 48:37-53. [PMID: 36100658 PMCID: PMC9700791 DOI: 10.1038/s41386-022-01438-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022]
Abstract
Endocannabinoids (eCBs) are lipid neuromodulators that suppress neurotransmitter release, reduce postsynaptic excitability, activate astrocyte signaling, and control cellular respiration. Here, we describe canonical and emerging eCB signaling modes and aim to link adaptations in these signaling systems to pathological states. Adaptations in eCB signaling systems have been identified in a variety of biobehavioral and physiological process relevant to neuropsychiatric disease states including stress-related disorders, epilepsy, developmental disorders, obesity, and substance use disorders. These insights have enhanced our understanding of the pathophysiology of neurological and psychiatric disorders and are contributing to the ongoing development of eCB-targeting therapeutics. We suggest future studies aimed at illuminating how adaptations in canonical as well as emerging cellular and synaptic modes of eCB signaling contribute to disease pathophysiology or resilience could further advance these novel treatment approaches.
Collapse
Affiliation(s)
| | - Farhana Yasmin
- Northwestern Center for Psychiatric Neuroscience, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Saptarnab Naskar
- Northwestern Center for Psychiatric Neuroscience, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sachin Patel
- Northwestern Center for Psychiatric Neuroscience, Chicago, IL, USA.
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
6
|
Molecular Alterations of the Endocannabinoid System in Psychiatric Disorders. Int J Mol Sci 2022; 23:ijms23094764. [PMID: 35563156 PMCID: PMC9104141 DOI: 10.3390/ijms23094764] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 02/07/2023] Open
Abstract
The therapeutic benefits of the current medications for patients with psychiatric disorders contrast with a great variety of adverse effects. The endocannabinoid system (ECS) components have gained high interest as potential new targets for treating psychiatry diseases because of their neuromodulator role, which is essential to understanding the regulation of many brain functions. This article reviewed the molecular alterations in ECS occurring in different psychiatric conditions. The methods used to identify alterations in the ECS were also described. We used a translational approach. The animal models reproducing some behavioral and/or neurochemical aspects of psychiatric disorders and the molecular alterations in clinical studies in post-mortem brain tissue or peripheral tissues were analyzed. This article reviewed the most relevant ECS changes in prevalent psychiatric diseases such as mood disorders, schizophrenia, autism, attentional deficit, eating disorders (ED), and addiction. The review concludes that clinical research studies are urgently needed for two different purposes: (1) To identify alterations of the ECS components potentially useful as new biomarkers relating to a specific disease or condition, and (2) to design new therapeutic targets based on the specific alterations found to improve the pharmacological treatment in psychiatry.
Collapse
|
7
|
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: 6] [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
|
8
|
Nguyen T, Gamage TF, Finlay DB, Decker AM, Langston TL, Barrus D, Glass M, Li JX, Kenakin TP, Zhang Y. Development of 3-(4-Chlorophenyl)-1-(phenethyl)urea Analogues as Allosteric Modulators of the Cannabinoid Type-1 Receptor: RTICBM-189 is Brain Penetrant and Attenuates Reinstatement of Cocaine-Seeking Behavior. J Med Chem 2021; 65:257-270. [PMID: 34929081 DOI: 10.1021/acs.jmedchem.1c01432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
We have shown that CB1 receptor negative allosteric modulators (NAMs) attenuated the reinstatement of cocaine-seeking behaviors in rats. In an effort to further define the structure-activity relationships and assess the druglike properties of the 3-(4-chlorophenyl)-1-(phenethyl)urea-based CB1 NAMs that we recently reported, we introduced substituents of different electronic properties and sizes to the phenethyl group and evaluated their potency in CB1 calcium mobilization, cAMP, and GTPγS assays. We found that 3-position substitutions such as Cl, F, and Me afforded enhanced CB1 potency, whereas 4-position analogues were generally less potent. The 3-chloro analogue (31, RTICBM-189) showed no activity at >50 protein targets and excellent brain permeation but relatively low metabolic stability in rat liver microsomes. Pharmacokinetic studies in rats confirmed the excellent brain exposure of 31 with a brain/plasma ratio Kp of 2.0. Importantly, intraperitoneal administration of 31 significantly and selectively attenuated the reinstatement of the cocaine-seeking behavior in rats without affecting locomotion.
Collapse
Affiliation(s)
- Thuy Nguyen
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Thomas F Gamage
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - David B Finlay
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Ann M Decker
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Tiffany L Langston
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Daniel Barrus
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Michelle Glass
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, the State University of New York, Buffalo, New York 14214, United States
| | - Terry P Kenakin
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
9
|
Chiu AS, Kang MC, Huerta Sanchez LL, Fabella AM, Holder KN, Barger BD, Elias KN, Shin CB, Jimenez Chavez CL, Kippin TE, Szumlinski KK. Preclinical evidence to support repurposing everolimus for craving reduction during protracted drug withdrawal. Neuropsychopharmacology 2021; 46:2090-2100. [PMID: 34188183 PMCID: PMC8505628 DOI: 10.1038/s41386-021-01064-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
Cue-elicited drug-craving is a cardinal feature of addiction that intensifies (incubates) during protracted withdrawal. In a rat model, these addiction-related behavioral pathologies are mediated, respectively, by time-dependent increases in PI3K/Akt1 signaling and reduced Group 1 metabotropic glutamate receptor (mGlu) expression, within the ventromedial prefrontal cortex (vmPFC). Herein, we examined the capacity of single oral dosing with everolimus, an FDA-approved inhibitor of the PI3K/Akt effector mTOR, to reduce incubated cocaine-craving and reverse incubation-associated changes in vmPFC kinase activity and mGlu expression. Rats were trained to lever-press for intravenous infusions of cocaine or delivery of sucrose pellets and then subjected to tests for cue-reinforced responding during early (3 days) or late (30-46 days) withdrawal. Rats were gavage-infused with everolimus (0-1.0 mg/kg), either prior to testing to examine for effects upon reinforcer-seeking behavior, or immediately following testing to probe effects upon the consolidation of extinction learning. Single oral dosing with everolimus dose-dependently blocked cocaine-seeking during late withdrawal and the effect lasted at least 24 h. No everolimus effects were observed for cue-elicited sucrose-seeking or cocaine-seeking in early withdrawal. In addition, everolimus treatment, following initial cue-testing, reduced subsequent cue hyper-responsivity exhibited observed during late withdrawal, arguing a facilitation of extinction memory consolidation. everolimus' "anti-incubation" effect was associated with a reversal of withdrawal-induced changes in indices of PI3K/Akt1/mTOR activity, as well as Homer protein and mGlu1/5 expression, within the prelimbic (PL) subregion of the prefrontal cortex. Our results indicate mTOR inhibition as a viable strategy for interrupting heightened cocaine-craving and facilitating addiction recovery during protracted withdrawal.
Collapse
Affiliation(s)
- Alvin S Chiu
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Matthew C Kang
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Laura L Huerta Sanchez
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Anne M Fabella
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Kalysta N Holder
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Brooke D Barger
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Kristina N Elias
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Christina B Shin
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - C Leonardo Jimenez Chavez
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Tod E Kippin
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Molecular, Developmental and Cell Biology and the Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
- Institute for Collaborative Biotechnologies, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA.
- Department of Molecular, Developmental and Cell Biology and the Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA.
| |
Collapse
|
10
|
Physical exercise rescues cocaine-evoked synaptic deficits in motor cortex. Mol Psychiatry 2021; 26:6187-6197. [PMID: 34686765 DOI: 10.1038/s41380-021-01336-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 09/24/2021] [Accepted: 10/01/2021] [Indexed: 02/07/2023]
Abstract
Drug exposure impairs cortical plasticity and motor learning, which underlies the reduced behavioral flexibility in drug addiction. Physical exercise has been used to prevent relapse in drug rehabilitation program. However, the potential benefits and molecular mechanisms of physical exercise on drug-evoked motor-cortical dysfunctions are unknown. Here we report that 1-week treadmill training restores cocaine-induced synaptic deficits, in the form of improved in vivo spine formation, synaptic transmission, and spontaneous activities of cortical pyramidal neurons, as well as motor-learning ability. The synaptic and behavioral benefits relied on de novo protein synthesis, which are directed by the activation of the mechanistic target of rapamycin (mTOR)-ribosomal protein S6 pathway. These findings establish synaptic functional restoration and mTOR signaling as the critical mechanism supporting physical exercise training in rehabilitating the addicted brain.
Collapse
|
11
|
Morris G, Walder K, Kloiber S, Amminger P, Berk M, Bortolasci CC, Maes M, Puri BK, Carvalho AF. The endocannabinoidome in neuropsychiatry: Opportunities and potential risks. Pharmacol Res 2021; 170:105729. [PMID: 34119623 DOI: 10.1016/j.phrs.2021.105729] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023]
Abstract
The endocannabinoid system (ECS) comprises two cognate endocannabinoid receptors referred to as CB1R and CB2R. ECS dysregulation is apparent in neurodegenerative/neuro-psychiatric disorders including but not limited to schizophrenia, major depressive disorder and potentially bipolar disorder. The aim of this paper is to review mechanisms whereby both receptors may interact with neuro-immune and neuro-oxidative pathways, which play a pathophysiological role in these disorders. CB1R is located in the presynaptic terminals of GABAergic, glutamatergic, cholinergic, noradrenergic and serotonergic neurons where it regulates the retrograde suppression of neurotransmission. CB1R plays a key role in long-term depression, and, to a lesser extent, long-term potentiation, thereby modulating synaptic transmission and mediating learning and memory. Optimal CB1R activity plays an essential neuroprotective role by providing a defense against the development of glutamate-mediated excitotoxicity, which is achieved, at least in part, by impeding AMPA-mediated increase in intracellular calcium overload and oxidative stress. Moreover, CB1R activity enables optimal neuron-glial communication and the function of the neurovascular unit. CB2R receptors are detected in peripheral immune cells and also in central nervous system regions including the striatum, basal ganglia, frontal cortex, hippocampus, amygdala as well as the ventral tegmental area. CB2R upregulation inhibits the presynaptic release of glutamate in several brain regions. CB2R activation also decreases neuroinflammation partly by mediating the transition from a predominantly neurotoxic "M1" microglial phenotype to a more neuroprotective "M2" phenotype. CB1R and CB2R are thus novel drug targets for the treatment of neuro-immune and neuro-oxidative disorders including schizophrenia and affective disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Stefan Kloiber
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 33 Ursula Franklin Street, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Paul Amminger
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| |
Collapse
|
12
|
Shapiro LP, Pitts EG, Li DC, Barbee BR, Hinton EA, Bassell GJ, Gross C, Gourley SL. The PI3-Kinase p110β Isoform Controls Severity of Cocaine-Induced Sequelae and Alters the Striatal Transcriptome. Biol Psychiatry 2021; 89:959-969. [PMID: 33773752 PMCID: PMC8202243 DOI: 10.1016/j.biopsych.2021.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/04/2021] [Accepted: 01/13/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The PI3-kinase (PI3K) complex is a well-validated target for mitigating cocaine-elicited sequelae, but pan-PI3K inhibitors are not viable long-term treatment options. The PI3K complex is composed of p110 catalytic and regulatory subunits, which can be individually manipulated for therapeutic purposes. However, this possibility has largely not been explored in behavioral contexts. METHODS Here, we inhibited PI3K p110β in the medial prefrontal cortex (mPFC) of cocaine-exposed mice. Behavioral models for studying relapse, sensitization, and decision-making biases were paired with protein quantification, RNA sequencing, and cell type-specific chemogenetic manipulation and RNA quantification to determine whether and how inhibiting PI3K p110β confers resilience to cocaine. RESULTS Viral-mediated PI3K p110β silencing reduced cue-induced reinstatement of cocaine seeking by half, blocked locomotor sensitization, and restored mPFC synaptic marker content after exposure to cocaine. Cocaine blocked the ability of mice to select actions based on their consequences, and p110β inhibition restored this ability. Silencing dopamine D2 receptor-expressing excitatory mPFC neurons mimicked cocaine, impairing goal-seeking behavior, and again, p110β inhibition restored goal-oriented action. We verified the presence of p110β in mPFC neurons projecting to the dorsal striatum and orbitofrontal cortex and found that inhibiting p110β in the mPFC altered the expression of functionally defined gene clusters within the dorsal striatum and not orbitofrontal cortex. CONCLUSIONS Subunit-selective PI3K silencing potently mitigates drug seeking, sensitization, and decision-making biases after exposure to cocaine. We suggest that inhibiting PI3K p110β provides neuroprotection against cocaine by triggering coordinated corticostriatal adaptations.
Collapse
Affiliation(s)
- Lauren P. Shapiro
- Graduate Program in Molecular and Systems Pharmacology, Emory University,Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center
| | - Elizabeth G. Pitts
- Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center,Graduate Program in Neuroscience, Emory University
| | - Dan C. Li
- Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center,Graduate Program in Neuroscience, Emory University
| | - Britton R. Barbee
- Graduate Program in Molecular and Systems Pharmacology, Emory University,Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center
| | - Elizabeth A. Hinton
- Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center,Graduate Program in Neuroscience, Emory University
| | - Gary J. Bassell
- Graduate Program in Neuroscience, Emory University,Department of Cell Biology, Emory University
| | - Christina Gross
- Division of Neurology, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine
| | - Shannon L. Gourley
- Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center,Graduate Program in Neuroscience, Emory University,Children’s Healthcare of Atlanta
| |
Collapse
|
13
|
Gasparyan A, Navarrete F, Rodríguez-Arias M, Miñarro J, Manzanares J. Cannabidiol Modulates Behavioural and Gene Expression Alterations Induced by Spontaneous Cocaine Withdrawal. Neurotherapeutics 2021; 18:615-623. [PMID: 33230690 PMCID: PMC8116402 DOI: 10.1007/s13311-020-00976-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 12/28/2022] Open
Abstract
The aim of this study was to evaluate the effects of cannabidiol (CBD) on the behavioural and gene expression changes in a new animal model of spontaneous cocaine withdrawal. For this purpose, male CD-1 mice were exposed to progressive increasing doses of cocaine for 12 days (15 to 60 mg/kg/day, i.p.), evaluating spontaneous cocaine withdrawal 6 h after the last cocaine administration. The effects of CBD (10, 20, and 40 mg/kg, i.p.) were evaluated on cocaine withdrawal-induced alterations in motor activity, somatic signs, and anxiety-like behaviour. Furthermore, gene expression changes in dopamine transporter (DAT) and tyrosine hydroxylase (TH) in the ventral tegmental area, and in cannabinoid receptors 1 (CNR1) and 2 (CNR2) in the nucleus accumbens, were analysed by real-time PCR. The results obtained in the study showed that mice exposed to the spontaneous cocaine withdrawal model presented increased motor activity, somatic withdrawal signs, and high anxiety-like behaviour. Interestingly, the administration of CBD normalized motor and somatic signs disturbances and induced an anxiolytic effect. Moreover, the administration of CBD blocked the increase of DAT and TH gene expression in mice exposed to the cocaine withdrawal, regulated the decrease of CNR1 and induced an additional upregulation of CNR2 gene expression. Thus, this model of spontaneous cocaine withdrawal induces clear behavioural and gene expression changes in mice. Interestingly, CBD alleviates these behavioural and gene expression alterations suggesting its potential for the management of cocaine withdrawal.
Collapse
Affiliation(s)
- Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550, Alicante, Spain
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550, Alicante, Spain
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Marta Rodríguez-Arias
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universidad de Valencia, Valencia, Spain
| | - José Miñarro
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universidad de Valencia, Valencia, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550, Alicante, Spain.
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.
| |
Collapse
|
14
|
Ucha M, Roura-Martínez D, Ambrosio E, Higuera-Matas A. The role of the mTOR pathway in models of drug-induced reward and the behavioural constituents of addiction. J Psychopharmacol 2020; 34:1176-1199. [PMID: 32854585 DOI: 10.1177/0269881120944159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Exposure to drugs of abuse induces neuroadaptations in critical nodes of the so-called reward systems that are thought to mediate the transition from controlled drug use to the compulsive drug-seeking that characterizes addictive disorders. These neural adaptations are likely to require protein synthesis, which is regulated, among others, by the mechanistic target of the rapamycin kinase (mTOR) signalling cascade. METHODS We have performed a narrative review of the literature available in PubMed about the involvement of the mTOR pathway in drug-reward and addiction-related phenomena. AIMS The aim of this study was to review the underlying architecture of this complex intracellular network and to discuss the alterations of its components that are evident after exposure to drugs of abuse. The aim was also to delineate the effects that manipulations of the mTOR network have on models of drug reward and on paradigms that recapitulate some of the psychological components of addiction. RESULTS There is evidence for the involvement of the mTOR pathway in the acute and rewarding effects of drugs of abuse, especially psychostimulants. However, the data regarding opiates are scarce. There is a need to use sophisticated animal models of addiction to ascertain the real role of the mTOR pathway in this pathology and not just in drug-mediated reward. The involvement of this pathway in behavioural addictions and impulsivity should also be studied in detail in the future. CONCLUSIONS Although there is a plethora of data about the modulation of mTOR by drugs of abuse, the involvement of this signalling pathway in addictive disorders requires further research.
Collapse
Affiliation(s)
- Marcos Ucha
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - David Roura-Martínez
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - Emilio Ambrosio
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - Alejandro Higuera-Matas
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| |
Collapse
|
15
|
Regulation of cannabinoid CB 1 and CB 2 receptors, neuroprotective mTOR and pro-apoptotic JNK1/2 kinases in postmortem prefrontal cortex of subjects with major depressive disorder. J Affect Disord 2020; 276:626-635. [PMID: 32871695 DOI: 10.1016/j.jad.2020.07.074] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/26/2020] [Accepted: 07/05/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Dysregulations of endocannabinoids and/or cannabinoid (CB) receptors have been implicated in the pathophysiology and treatment of major depressive disorder (MDD). METHODS CB1 and CB2 receptors, neuroprotective mTOR (mechanistic target of rapamycin) and pro-apoptotic JNK1/2 (c-Jun-N-terminal kinases) were quantified by immunoblotting in postmortem prefrontal cortex of MDD and controls, and further compared in antidepressant (AD)-free and AD-treated subjects. Neuroplastic proteins (PSD-95, Arc, spinophilin) were quantified in MDD brains. RESULTS Total cortical CB1 glycosylated (≈54/64 kDa) receptor was increased in MDD (+20%, n=23, p=0.02) when compared with controls (100%, n=19). This CB1 receptor upregulation was quantified in AD-treated (+23%, n=14, p=0.02) but not in AD-free (+14%, n=9, p=0.34) MDD subjects. In the same MDD cortical samples, CB2 glycosylated (≈45 kDa) receptor was unaltered (all MDD: +11%, n=23, p=0.10; AD-free: +12%, n=9, p=0.31; AD-treated: +10%, n=14, p=0.23). In MDD, mTOR activity (p-Ser2448 TOR/t-TOR) was increased (all MDD: +29%, n=18, p=0.002; AD-free: +33%, n=8, p=0.03; AD-treated: +25%, n=10, p=0.04). In contrast, JNK1/2 activity (p-Thr183/Tyr185/t-JNK) was unaltered in MDD subjects. Cortical PSD-95, Arc, and spinophilin contents were unchanged in MDD. LIMITATIONS A relative limited sample size. Some MDD subjects had been treated with a variety of ADs. The results must be understood in the context of suicide victims with MDD. CONCLUSIONS The upregulation of CB1 receptor density, but not that of CB2 receptor, as well as the increased mTOR activity in PFC/BA9 of subjects with MDD (AD-free/treated) support their contributions in the complex pathophysiology of MDD and in the molecular mechanisms of antidepressant drugs.
Collapse
|
16
|
Wang R, Hausknecht KA, Gancarz-Kausch AM, Oubraim S, Shen RY, Haj-Dahmane S. Cocaine self-administration abolishes endocannabinoid-mediated long-term depression of glutamatergic synapses in the ventral tegmental area. Eur J Neurosci 2020; 52:4517-4524. [PMID: 32959420 DOI: 10.1111/ejn.14980] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/24/2020] [Accepted: 09/07/2020] [Indexed: 01/26/2023]
Abstract
Drugs of abuse, including cocaine, alter the mechanisms underpinning synaptic plasticity, including long-term potentiation of glutamatergic synapses in the mesolimbic system. These effects are thought to underlie addictive behaviors. In the ventral tegmental area (VTA), glutamatergic synapses also exhibit long-term depression (LTD), a type of plasticity that weakens synaptic strength. This form of synaptic plasticity is induced by low-frequency stimulation and mediated by endocannabinoid (eCB) signaling, which also modulates addictive behaviors. However, it remains unknown whether eCB-LTD in the VTA could be altered by cocaine use. Therefore, the goal of the present study was to examine the impact of cocaine self-administration on eCB-LTD of glutamatergic synapses onto VTA dopaminergic (DA) neurons. To that end, male rats underwent cocaine (0.75 mg/kg/infusion) or saline self-administration under the fixed ratio 1 schedule for 6-9 days. One day after the last self-administration session, the magnitude of eCB-LTD was examined using ex vivo whole-cell recordings of putative VTA DA neurons from naïve rats and rats with saline or cocaine self-administration. The results revealed that cocaine self-administration abolished eCB-LTD. The cocaine-induced blockade of eCB-LTD in the VTA was mediated by an impaired function of presynaptic CB1 receptors. Collectively, these findings indicate that cocaine exposure blunts eCB-mediated synaptic plasticity in midbrain DA neurons. This effect could be one of the cellular mechanisms that mediate, at least in part, addictive behaviors.
Collapse
Affiliation(s)
- Ruixiang Wang
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Kathryn A Hausknecht
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Amy M Gancarz-Kausch
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.,Department of Psychology, California State University Bakersfield, Bakersfield, CA, USA
| | - Saida Oubraim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
17
|
Phosphoproteomic Analysis of the Amygdala Response to Adolescent Glucocorticoid Exposure Reveals G-Protein Coupled Receptor Kinase 2 as a Target for Reducing Motivation for Alcohol. Proteomes 2018; 6:proteomes6040041. [PMID: 30322021 PMCID: PMC6313880 DOI: 10.3390/proteomes6040041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/28/2018] [Accepted: 10/09/2018] [Indexed: 01/19/2023] Open
Abstract
Early life stress is associated with risk for developing alcohol use disorders (AUDs) in adulthood. Though the neurobiological mechanisms underlying this vulnerability are not well understood, evidence suggests that aberrant glucocorticoid and noradrenergic system functioning play a role. The present study investigated the long-term consequences of chronic exposure to elevated glucocorticoids during adolescence on the risk of increased alcohol-motivated behavior, and on amygdalar function in adulthood. A discovery-based analysis of the amygdalar phosphoproteome using mass spectrometry was employed, to identify changes in function. Adolescent corticosterone (CORT) exposure increased alcohol, but not sucrose, self-administration, and enhanced stress-induced reinstatement with yohimbine in adulthood. Phosphoproteomic analysis indicated that the amygdala phosphoproteome was significantly altered by adolescent CORT exposure, generating a list of potential novel mechanisms involved in the risk of alcohol drinking. In particular, increased phosphorylation at serines 296–299 on the α2A adrenergic receptor (α2AAR), mediated by the G-protein coupled receptor kinase 2 (GRK2), was evident after adolescent CORT exposure. We found that intra-amygdala infusion of a peptidergic GRK2 inhibitor reduced alcohol seeking, as measured by progressive ratio and stress reinstatement tests, and induced by the α2AAR antagonist yohimbine. These results suggest that GRK2 represents a novel target for treating stress-induced motivation for alcohol which may counteract alterations in brain function induced by adolescent stress exposure.
Collapse
|
18
|
Huang SH, Wu WR, Lee LM, Huang PR, Chen JC. mTOR signaling in the nucleus accumbens mediates behavioral sensitization to methamphetamine. Prog Neuropsychopharmacol Biol Psychiatry 2018; 86:331-339. [PMID: 29574227 DOI: 10.1016/j.pnpbp.2018.03.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 01/01/2023]
Abstract
Chronic psychostimulant treatment in rodents readily produces behavioral sensitization, which reflects altered brain function in response to repeated drug exposure. Numerous morphological and biochemical investigations implicate altered neural plasticity in striatal medium spiny neurons (MSNs) as an essential component in behavioral sensitization. The mammalian target of the rapamycin (mTOR) signaling pathway, a key regulator of synaptic neuroplasticity, in the ventral striatum of methamphetamine (METH) -sensitized mice was investigated to determine if a link exists with the development of METH sensitization. Behaviorally, METH-sensitized mice possessed increased levels of phosphorylated mTOR/S2448 and its down-stream regulator p70S6K and pS6 in the ventral striatum. Systemic treatment with rapamycin, a specific mTOR inhibitor, coincident with a daily METH injection suppressed the induction of METH sensitization and reduced the number of dendritic spines in the shell and core of the nucleus accumbens. The infusion of lentivirus-expressing mTOR-shRNA into the shell region of the nucleus accumbens inhibited the induction of behavioral sensitization to METH, which was comparable to the effect of rapamycin. These results suggest that mTORC1-mediated signaling in the nucleus accumbens mediates the development of behavioral sensitization to METH.
Collapse
Affiliation(s)
- Shin-Han Huang
- Department of Physiology and Pharmacology, Graduate Institute of Biomedical Sciences, School of Medicine, Chang-Gung University, Taiwan
| | - Wan-Rong Wu
- Department of Physiology and Pharmacology, Graduate Institute of Biomedical Sciences, School of Medicine, Chang-Gung University, Taiwan
| | - Li-Ming Lee
- Department of Biomedical Sciences, School of Medicine, Chang-Gung University, Taiwan
| | - Pei-Rong Huang
- Center for Molecular and Clinical Immunology, Chang-Gung University, Taiwan
| | - Jin-Chung Chen
- Department of Physiology and Pharmacology, Graduate Institute of Biomedical Sciences, School of Medicine, Chang-Gung University, Taiwan; Healthy Aging Research Center, Chang-Gung University, Taiwan; Neuroscience Research Center, Chang-Gung Memorial Hospital, Linkou, Taiwan; Chang-Gung Memorial Hospital, Keelung, Taiwan.
| |
Collapse
|
19
|
Bystrowska B, Frankowska M, Smaga I, Pomierny-Chamioło L, Filip M. Effects of Cocaine Self-Administration and Its Extinction on the Rat Brain Cannabinoid CB1 and CB2 Receptors. Neurotox Res 2018; 34:547-558. [PMID: 29754307 PMCID: PMC6154179 DOI: 10.1007/s12640-018-9910-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 04/23/2018] [Accepted: 05/01/2018] [Indexed: 12/18/2022]
Abstract
The aim of this study was to evaluate changes in the expression of cannabinoid type 1 (CB1) and 2 (CB2) receptor proteins in several brain regions in rats undergoing cocaine self-administration and extinction training. We used a triad-yoked procedure to distinguish between the motivational and pharmacological effects of cocaine. Using immunohistochemistry, we observed a significant decrease in CB1 receptor expression in the prefrontal cortex, dorsal striatum, and the basolateral and basomedial amygdala following cocaine (0.5 mg/kg/infusion) self-administration. Increased CB1 receptor expression in the ventral tegmental area in rats with previous cocaine exposure was also found. Following cocaine abstinence after 10 days of extinction training, we detected increases in the expression of CB1 receptors in the substantia nigra in both cocaine groups and in the subregions of the amygdala for only the yoked cocaine controls, while any method of cocaine exposure resulted in a decrease in CB2 receptor expression in the prefrontal cortex (p < 0.01), nucleus accumbens (p < 0.01), and medial globus pallidus (p < 0.01). Our findings further support the idea that the eCB system and CB1 receptors are involved in cocaine-reinforced behaviors. Moreover, we detected a cocaine-evoked adaptation in CB2 receptors in the amygdala, prefrontal cortex, and globus pallidus.
Collapse
Affiliation(s)
- Beata Bystrowska
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| | - Małgorzata Frankowska
- Department of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Irena Smaga
- Department of Internal Medicine, Jagiellonian University Medical College, Skawińska 8, 31-066, Kraków, Poland
| | - Lucyna Pomierny-Chamioło
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Małgorzata Filip
- Department of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| |
Collapse
|
20
|
Cooper A, Singh S, Hook S, Tyndall JDA, Vernall AJ. Chemical Tools for Studying Lipid-Binding Class A G Protein-Coupled Receptors. Pharmacol Rev 2017; 69:316-353. [PMID: 28655732 DOI: 10.1124/pr.116.013243] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 05/15/2017] [Indexed: 12/16/2022] Open
Abstract
Cannabinoid, free fatty acid, lysophosphatidic acid, sphingosine 1-phosphate, prostanoid, leukotriene, bile acid, and platelet-activating factor receptor families are class A G protein-coupled receptors with endogenous lipid ligands. Pharmacological tools are crucial for studying these receptors and addressing the many unanswered questions surrounding expression of these receptors in normal and diseased tissues. An inherent challenge for developing tools for these lipid receptors is balancing the often lipophilic requirements of the receptor-binding pharmacophore with favorable physicochemical properties to optimize highly specific binding. In this study, we review the radioligands, fluorescent ligands, covalent ligands, and antibodies that have been used to study these lipid-binding receptors. For each tool type, the characteristics and design rationale along with in vitro and in vivo applications are detailed.
Collapse
Affiliation(s)
- Anna Cooper
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Sameek Singh
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | | | | |
Collapse
|
21
|
Castilla-Ortega E, Ladrón de Guevara-Miranda D, Serrano A, Pavón FJ, Suárez J, Rodríguez de Fonseca F, Santín LJ. The impact of cocaine on adult hippocampal neurogenesis: Potential neurobiological mechanisms and contributions to maladaptive cognition in cocaine addiction disorder. Biochem Pharmacol 2017; 141:100-117. [DOI: 10.1016/j.bcp.2017.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022]
|
22
|
Butelman ER, Bacciardi S, Maremmani AGI, Darst-Campbell M, Correa da Rosa J, Kreek MJ. Can a rapid measure of self-exposure to drugs of abuse provide dimensional information on depression comorbidity? Am J Addict 2017; 26:632-639. [PMID: 28654734 DOI: 10.1111/ajad.12578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/28/2017] [Accepted: 06/04/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Addictions to heroin or to cocaine are associated with substantial psychiatric comorbidity, including depression. Poly-drug self-exposure (eg, to heroin, cocaine, cannabis, or alcohol) is also common, and may further affect depression comorbidity. METHODS This case-control study examined the relationship of exposure to the above drugs and depression comorbidity. Participants were recruited from methadone maintenance clinics, and from the community. Adult male and female participants (n = 1,201) were ascertained consecutively by experienced licensed clinicians. The instruments used were the SCID-I, and Kreek-McHugh-Schluger-Kellogg (KMSK) scales, which provide a rapid dimensional measure of maximal lifetime self-exposure to each of the above drugs. This measure ranges from no exposure to high unit dose, high frequency, and long duration of exposure. RESULTS A multiple logistic regression with stepwise variable selection revealed that increasing exposure to heroin or to cocaine was associated greater odds of depression, with all cases and controls combined. In cases with an opioid dependence diagnosis, increasing cocaine exposure was associated with a further increase in odds of depression. However, in cases with a cocaine dependence diagnosis, increasing exposure to either cannabis or alcohol, as well as heroin, was associated with a further increase in odds of depression. DISCUSSION AND CONCLUSIONS This dimensional analysis of exposure to specific drugs provides insights on depression comorbidity with addictive diseases, and the impact of poly-drug exposure. SCIENTIFIC SIGNIFICANCE A rapid analysis of exposure to drugs of abuse reveals how specific patterns of drug and poly-drug exposure are associated with increasing odds of depression. This approach detected quantitatively how different patterns of poly-drug exposure can result in increased odds of depression comorbidity, in cases diagnosed with opioid versus cocaine dependence. (Am J Addict 2017;26:632-639).
Collapse
Affiliation(s)
- Eduardo Roque Butelman
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Silvia Bacciardi
- "VP Dole" Dual Diagnosis Unit, Santa Chiara University Hospital of Pisa, Pisa, Italy
| | | | - Maya Darst-Campbell
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Joel Correa da Rosa
- Center for Clinical and Translational Science, The Rockefeller University Hospital, New York, New York
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| |
Collapse
|
23
|
Smaga I, Jastrzębska J, Zaniewska M, Bystrowska B, Gawliński D, Faron-Górecka A, Broniowska Ż, Miszkiel J, Filip M. Changes in the Brain Endocannabinoid System in Rat Models of Depression. Neurotox Res 2017; 31:421-435. [PMID: 28247204 PMCID: PMC5360820 DOI: 10.1007/s12640-017-9708-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 02/02/2017] [Accepted: 02/07/2017] [Indexed: 12/23/2022]
Abstract
A growing body of evidence implicates the endocannabinoid (eCB) system in the pathophysiology of depression. The aim of this study was to investigate the influence of changes in the eCB system, such as levels of neuromodulators, eCB synthesizing and degrading enzymes, and cannabinoid (CB) receptors, in different brain structures in animal models of depression using behavioral and biochemical analyses. Both models used, i.e., bulbectomized (OBX) and Wistar Kyoto (WKY) rats, were characterized at the behavioral level by increased immobility time. In the OBX rats, anandamide (AEA) levels were decreased in the prefrontal cortex, hippocampus, and striatum and increased in the nucleus accumbens, while 2-arachidonoylglycerol (2-AG) levels were increased in the prefrontal cortex and decreased in the nucleus accumbens with parallel changes in the expression of eCB metabolizing enzymes in several structures. It was also observed that CB1 receptor expression decreased in the hippocampus, dorsal striatum, and nucleus accumbens, and CB2 receptor expression decreased in the prefrontal cortex and hippocampus. In WKY rats, the levels of eCBs were reduced in the prefrontal cortex (2-AG) and dorsal striatum (AEA) and increased in the prefrontal cortex (AEA) with different changes in the expression of eCB metabolizing enzymes, while the CB1 receptor density was increased in several brain regions. These findings suggest that dysregulation in the eCB system is implicated in the pathogenesis of depression, although neurochemical changes were linked to the particular brain structure and the factor inducing depression (surgical removal of the olfactory bulbs vs. genetic modulation).
Collapse
Affiliation(s)
- Irena Smaga
- Department of Toxicology, Faculty of Pharmacy, College of Medicum, Jagiellonian University, Medyczna 9, PL 30-688, Kraków, Poland
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343, Kraków, Poland
| | - Joanna Jastrzębska
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343, Kraków, Poland
| | - Magdalena Zaniewska
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343, Kraków, Poland
| | - Beata Bystrowska
- Department of Toxicology, Faculty of Pharmacy, College of Medicum, Jagiellonian University, Medyczna 9, PL 30-688, Kraków, Poland
| | - Dawid Gawliński
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343, Kraków, Poland
| | - Agata Faron-Górecka
- Laboratory of Biochemical Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343, Kraków, Poland
| | - Żaneta Broniowska
- Department of Toxicology, Faculty of Pharmacy, College of Medicum, Jagiellonian University, Medyczna 9, PL 30-688, Kraków, Poland
| | - Joanna Miszkiel
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343, Kraków, Poland
| | - Małgorzata Filip
- Department of Toxicology, Faculty of Pharmacy, College of Medicum, Jagiellonian University, Medyczna 9, PL 30-688, Kraków, Poland.
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343, Kraków, Poland.
| |
Collapse
|
24
|
Salort G, Álvaro-Bartolomé M, García-Sevilla JA. Regulation of cannabinoid CB 2 receptor constitutive activity in vivo: repeated treatments with inverse agonists reverse the acute activation of JNK and associated apoptotic signaling in mouse brain. Psychopharmacology (Berl) 2017; 234:925-941. [PMID: 28127623 DOI: 10.1007/s00213-017-4537-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/07/2017] [Indexed: 01/29/2023]
Abstract
RATIONALE CB2 receptors express constitutive activity and inverse agonists regulate receptor basal activity, which might be involved in death mechanisms. This study assessed the effects of a selective CB2 agonist (JWH133) and different CB2 inverse agonists (AM630, JTE907, raloxifene) on death pathways in brain. OBJECTIVES The acute (JWH13) and the acute/chronic effects (AM630, JTE907, raloxifene) of CB2 ligands regulating pro-apoptotic c-Jun NH2-terminal kinase (p-JNK/JNK ratio) and associated signaling of extrinsic (Fas receptor, Fas-Associated death domain protein, FADD) and intrinsic (Bax, cytochrome c) death pathways (nuclear poly (ADP-ribose) polymerase PARP) were investigated in mouse brain. METHODS Mice were treated with CB2 drugs and target protein contents were assessed by western blot analysis. RESULTS JWH133 reduced cortical JNK (-27-45%) whereas AM630 acutely increased JNK in cortex (+61-148%), cerebellum (+34-40%), and striatum (+33-42%). JTE907 and raloxifene also increased cortical JNK (+31%-57%). Acute AM630, but not JWH133, increased cortical FADD, Bax, cytochrome c, and PARP cleavage. Repeated treatments with the three CB2 inverse agonists were associated with a reversal of the acute effects resulting in decreases in cortical JNK (AM630: -36%; JTE907: -25%; raloxifene: -11%). Chronic treatments also induced a reversal with down-regulation (AM630) or only tolerance (JTE907 and raloxifene) on other apoptotic markers (FADD, Bax, cytochrome c, PARP). CONCLUSIONS AM630 and JTE907 are CB2 protean ligands. Thus, chronic inverse agonists abolished CB2 constitutive activity and then the ligands behaved as agonists reducing (like JWH133) JNK activity. Acute and chronic treatments with CB2 inverse agonists regulate in opposite directions brain death markers.
Collapse
Affiliation(s)
- Glòria Salort
- Laboratori de Neurofarmacologia, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS/IdisPa), Universitat de les Illes Balears, Cra. Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain.,Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - María Álvaro-Bartolomé
- Laboratori de Neurofarmacologia, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS/IdisPa), Universitat de les Illes Balears, Cra. Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain.,Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - Jesús A García-Sevilla
- Laboratori de Neurofarmacologia, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS/IdisPa), Universitat de les Illes Balears, Cra. Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain. .,Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain.
| |
Collapse
|
25
|
Johnson KA, Lovinger DM. Presynaptic G Protein-Coupled Receptors: Gatekeepers of Addiction? Front Cell Neurosci 2016; 10:264. [PMID: 27891077 PMCID: PMC5104741 DOI: 10.3389/fncel.2016.00264] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 10/31/2016] [Indexed: 12/21/2022] Open
Abstract
Drug abuse and addiction cause widespread social and public health problems, and the neurobiology underlying drug actions and drug use and abuse is an area of intensive research. Drugs of abuse alter synaptic transmission, and these actions contribute to acute intoxication as well as the chronic effects of abused substances. Transmission at most mammalian synapses involves neurotransmitter activation of two receptor subtypes, ligand-gated ion channels that mediate fast synaptic responses and G protein-coupled receptors (GPCRs) that have slower neuromodulatory actions. The GPCRs represent a large proportion of neurotransmitter receptors involved in almost all facets of nervous system function. In addition, these receptors are targets for many pharmacotherapeutic agents. Drugs of abuse directly or indirectly affect neuromodulation mediated by GPCRs, with important consequences for intoxication, drug taking and responses to prolonged drug exposure, withdrawal and addiction. Among the GPCRs are several subtypes involved in presynaptic inhibition, most of which are coupled to the Gi/o class of G protein. There is increasing evidence that these presynaptic Gi/o-coupled GPCRs have important roles in the actions of drugs of abuse, as well as behaviors related to these drugs. This topic will be reviewed, with particular emphasis on receptors for three neurotransmitters, Dopamine (DA; D1- and D2-like receptors), Endocannabinoids (eCBs; CB1 receptors) and glutamate (group II metabotropic glutamate (mGlu) receptors). The focus is on recent evidence from laboratory animal models (and some evidence in humans) implicating these receptors in the acute and chronic effects of numerous abused drugs, as well as in the control of drug seeking and taking. The ability of drugs targeting these receptors to modify drug seeking behavior has raised the possibility of using compounds targeting these receptors for addiction pharmacotherapy. This topic is also discussed, with emphasis on development of mGlu2 positive allosteric modulators (PAMs).
Collapse
Affiliation(s)
- Kari A. Johnson
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, USA
| | - David M. Lovinger
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, USA
| |
Collapse
|
26
|
Luo YX, Han H, Shao J, Gao Y, Yin X, Zhu WL, Han Y, Shi HS. mTOR signalling in the nucleus accumbens shell is critical for augmented effect of TFF3 on behavioural response to cocaine. Sci Rep 2016; 6:27895. [PMID: 27282818 PMCID: PMC4901260 DOI: 10.1038/srep27895] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/26/2016] [Indexed: 12/25/2022] Open
Abstract
Neuropeptides play important roles in modulating the rewarding value of abused drugs. Trefoil factor 3 (TFF3) was recently reported to modulate withdrawal syndrome of morphine, but the effects of TFF3 on the cocaine-induced behavioral changes are still elusive. In the present study, cocaine-induced hyperlocomotion and conditioned place preference (CPP) rat paradigms were provided to investigate the role of TFF3 in the reward response to cocaine. High-performance liquid chromatography (HPLC) analysis was used to analyse the dopamine concentration. The results showed that systemic TFF3 administration (0.1 mg/kg i.p.) significantly augmented cocaine- induced hyperlocomotion and CPP formation, without any effects on locomotor activity and aversive or rewarding effects per se. TFF3 significantly augmented the increment of the dopamine concentration in the NAc and the activity of the mTOR signalling pathway induced by acute cocaine exposure (10 mg/kg, i.p.) in the NAc shell, but not the core. The Intra-NAc shell infusion of rapamycin blocked TFF3-induced hyperactivity in cocaine-treatment rats. These findings indicated that TFF3 could potentiate behavioural response to cocaine, which may be associated with regulating dopamine concentration. Furthermore, the findings indicated that mTOR signalling pathway in the NAc shell is important for TFF3-induced enhancement on the cocaine-induced behavioral changes.
Collapse
Affiliation(s)
- Yi-Xiao Luo
- Department of Pharmacology, Medical School of Hunan Normal University, Changsha 410013, China
| | - Hua Han
- Department of gynecology and obstetrics, Hebei General Hospital, Shijiazhuang 050051, China
| | - Juan Shao
- Department of Senile Disease, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Yuan Gao
- Department of Biochemistry and Molecular Biology, College of basic medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Xi Yin
- Department of Functional region of Diagnosis, Hebei Medical University Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Wei-Li Zhu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Ying Han
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Hai-Shui Shi
- Department of Biochemistry and Molecular Biology, College of basic medicine, Hebei Medical University, Shijiazhuang 050017, China.,National Institute on Drug Dependence, Peking University, Beijing 100191, China
| |
Collapse
|
27
|
García-Pardo MP, Roger-Sanchez C, Rodríguez-Arias M, Miñarro J, Aguilar MA. Pharmacological modulation of protein kinases as a new approach to treat addiction to cocaine and opiates. Eur J Pharmacol 2016; 781:10-24. [DOI: 10.1016/j.ejphar.2016.03.065] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/28/2016] [Accepted: 03/31/2016] [Indexed: 12/13/2022]
|
28
|
Rubio M, Valdeolivas S, Piscitelli F, Verde R, Satta V, Barroso E, Montolio M, Aras LM, Di Marzo V, Sagredo O, Fernández-Ruiz J. Analysis of endocannabinoid signaling elements and related proteins in lymphocytes of patients with Dravet syndrome. Pharmacol Res Perspect 2016; 4:e00220. [PMID: 27069631 PMCID: PMC4804326 DOI: 10.1002/prp2.220] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/20/2016] [Indexed: 11/09/2022] Open
Abstract
Cannabidiol (CBD) reduces seizures in childhood epilepsy syndromes including Dravet syndrome (DS). A formulation of CBD has obtained orphan drug designation for these syndromes and clinical trials are currently underway. The mechanism responsible for CBD effects is not known, although it could involve targets sensitive to CBD in other neurological disorders. We believe of interest to investigate whether these potential targets are altered in DS, in particular whether the endocannabinoid system is dysregulated. To this end, lymphocytes from patients and controls were used for analysis of gene expression of transmitter receptors and transporters, ion channels, and enzymes associated with CBD effects, as well as endocannabinoid genes. Plasma endocannabinoid levels were also analyzed. There were no differences between DS patients and controls in most of the CBD targets analyzed, except an increase in the voltage-dependent calcium channel α-1h subunit. We also found that cannabinoid type-2 (CB 2) receptor gene expression was elevated in DS patients, with no changes in other endocannabinoid-related receptors and enzymes, as well as in plasma levels of endocannabinoids. Such elevation was paralleled by an increase in CD70, a marker of lymphocyte activation, and certain trends in inflammation-related proteins (e.g., peroxisome proliferator-activated receptor-γ receptors, cytokines). In conclusion, together with changes in the voltage-dependent calcium channel α-1h subunit, we found an upregulation of CB 2 receptors, associated with an activation of lymphocytes and changes in inflammation-related genes, in DS patients. Such changes were also reported in inflammatory disorders and may indirectly support the occurrence of a potential dysregulation of the endocannabinoid system in the brain.
Collapse
Affiliation(s)
- Marta Rubio
- Departamento de Bioquímica y Biología Molecular Facultad de Medicina Instituto Universitario de Investigación en Neuroquímica Universidad Complutense Madrid Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Madrid Spain
| | - Sara Valdeolivas
- Departamento de Bioquímica y Biología Molecular Facultad de Medicina Instituto Universitario de Investigación en Neuroquímica Universidad Complutense Madrid Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Madrid Spain
| | - Fabiana Piscitelli
- Endocannabinoid Research Group Institute of Biomolecular Chemistry Consiglio Nazionale delle Ricerche Pozzuoli, Naples Italy
| | - Roberta Verde
- Endocannabinoid Research Group Institute of Biomolecular Chemistry Consiglio Nazionale delle Ricerche Pozzuoli, Naples Italy
| | - Valentina Satta
- Departamento de Bioquímica y Biología Molecular Facultad de Medicina Instituto Universitario de Investigación en Neuroquímica Universidad Complutense Madrid Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Madrid Spain
| | - Eva Barroso
- Instituto de Genética Médica y Molecular (INGEMM)Hospital Universitario La Paz Universidad Autónoma de MadridIdi PAZ Madrid Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER) Instituto de Salud Carlos III Madrid Spain
| | - Marisol Montolio
- Dravet Syndrome Foundation Madrid Spain; Departamento de Biología Celular Facultad de Biología Universidad de Barcelona Barcelona Spain
| | - Luis Miguel Aras
- Dravet Syndrome Foundation Madrid Spain; Servicio Navarro de Salud Osasunbidea Estella Spain
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group Institute of Biomolecular Chemistry Consiglio Nazionale delle Ricerche Pozzuoli, Naples Italy
| | - Onintza Sagredo
- Departamento de Bioquímica y Biología Molecular Facultad de Medicina Instituto Universitario de Investigación en Neuroquímica Universidad Complutense Madrid Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Madrid Spain
| | - Javier Fernández-Ruiz
- Departamento de Bioquímica y Biología Molecular Facultad de Medicina Instituto Universitario de Investigación en Neuroquímica Universidad Complutense Madrid Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Madrid Spain
| |
Collapse
|
29
|
García-Cabrerizo R, García-Fuster MJ. Opposite regulation of cannabinoid CB1 and CB2 receptors in the prefrontal cortex of rats treated with cocaine during adolescence. Neurosci Lett 2016; 615:60-5. [PMID: 26797579 DOI: 10.1016/j.neulet.2016.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 12/21/2015] [Accepted: 01/13/2016] [Indexed: 12/09/2022]
Abstract
The endocannabinoid system is implicated in the neurobiology of cocaine addiction, although it is not clear how cocaine regulates brain CB1 and CB2 receptors, especially during adolescence, a critical moment for shaping adult response to drug use. This study evaluated CB1 and CB2 protein levels in prefrontal cortex (PFC) and hippocampus (HC) by western blot analysis with specific and validated antibodies: (1) basally during adolescence (post-natal day PND 40, PND 47, PND 54), (2) by a sensitizing regimen of cocaine (15mg/kg, 7 days, i.p.) during different windows of adolescence vulnerability (PND 33-39, PND 40-46, PND 47-53), and (3) following repeated cocaine administration during adolescence (PND 33-39) in adulthood (PND 64). The results demonstrated a dynamic and opposite basal modulation of CB1 and CB2 receptors in PFC and HC during adolescence. CB1 receptor levels were increased while CB2 receptors were decreased as compared to adulthood with asymptotes values around mid adolescence (PND 47) both in PFC (CB1: +45±22, p<0.05; CB2: -24±6%, p<0.05) and HC (CB1: +53±23, p<0.05; CB2: -20±8%, p<0.05). Interestingly, cocaine only altered CB1 (+55±10%, p<0.05) and CB2 (-25±10%, p<0.05) receptors when administered during early adolescence and only in PFC. However, the changes observed in PFC by repeated cocaine administration in adolescence were transient and did not endure into adulthood. These results identified a period of vulnerability during adolescence at which cocaine dysregulated the content of CB receptors in PFC, suggesting an opposite role for these receptors in the effects mediated by cocaine.
Collapse
Affiliation(s)
- Rubén García-Cabrerizo
- Neurobiology of Drug Abuse Group, IUNICS/IdISPa, University of the Balearic Islands, Palma de Mallorca, Spain; Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - M Julia García-Fuster
- Neurobiology of Drug Abuse Group, IUNICS/IdISPa, University of the Balearic Islands, Palma de Mallorca, Spain; Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain.
| |
Collapse
|
30
|
Keller B, García-Sevilla JA. Immunodetection and subcellular distribution of imidazoline receptor proteins with three antibodies in mouse and human brains: Effects of treatments with I1- and I2-imidazoline drugs. J Psychopharmacol 2015; 29:996-1012. [PMID: 26038110 DOI: 10.1177/0269881115586936] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Various imidazoline receptor (IR) proteins have been proposed to mediate the effects of selective I1- and I2-IR drugs. However, the association of these IR-binding proteins with classic I1- and I2-radioligand binding sites remains somewhat controversial. In this study, three IR antibodies (anti-NISCH and anti-nischarin for I1-IRs; and anti-IRBP for I1/I2-IRs) were used to immunodetect, characterize and compare IR protein patterns in brain (mouse and human; total homogenate, subcellular fractionation, grey and white matter) and some cell systems (neurones, astrocytes, human platelets). Various immunoreactive IRs (specific molecular weight bands coincidently detected with the different antibodies) were related to I1-IR (167 kDa, 105/115 kDa and 85 kDa proteins) or I2-IR (66 kDa, 45 kDa and 30 kDa proteins) types. The biochemical characterization of cortical 167 kDa protein, localized in the membrane/cytosol but not in the nucleus, indicated that this I1-IR also forms part of higher order nischarin-related complexes. The contents of I1-IR (167 kDa, 105/115 kDa, and 85 kDa) proteins in mouse brain cortex were upregulated by treatment with I1-drugs (moxonidine, efaroxan) but not with I2-drugs (BU-224, LSL 61122). Conversely, the contents of I2-IR (66 kDa, 45 kDa and 30 kDa) proteins in mouse brain cortex were modulated by treatment with I2-drugs (decreases after BU-224 and LSL 61122, and increases after idazoxan) but not with I1-drugs (with the exception of moxonidine). These findings further indicate that brain immunoreactive IR proteins exist in multiple forms that can be grouped in the already known I1- and I2-IR types, which are expressed both in neurones and astrocytes.
Collapse
Affiliation(s)
- Benjamin Keller
- Laboratori de Neurofarmacologia, IUNICS-IdISPa, Universitat de les Illes Balears, Palma de Mallorca, Spain and Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - Jesús A García-Sevilla
- Laboratori de Neurofarmacologia, IUNICS-IdISPa, Universitat de les Illes Balears, Palma de Mallorca, Spain and Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| |
Collapse
|
31
|
Hitchcock LN, Lattal KM. Histone-mediated epigenetics in addiction. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 128:51-87. [PMID: 25410541 DOI: 10.1016/b978-0-12-800977-2.00003-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Many of the brain regions, neurotransmitter systems, and behavioral changes that occur after occasional drug use in healthy subjects and after chronic drug abuse in addicted patients are well characterized. An emerging literature suggests that epigenetic processes, those processes that regulate the accessibility of DNA to regulatory proteins within the nucleus, are keys to how addiction develops and how it may be treated. Investigations of the regulation of chromatin, the organizational system of DNA, by histone modification are leading to a new understanding of the cellular and behavioral alterations that occur after drug use. We will describe how, when, and where histone tails are modified and how some of the most recognized histone regulation patterns are involved in the cycle of addiction, including initial and chronic drug intake, withdrawal, abstinence, and relapse. Finally, we consider how an approach that targets histone modifications may promote successful treatment.
Collapse
Affiliation(s)
- Leah N Hitchcock
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
32
|
García-Cabrerizo R, García-Fuster MJ. Chronic MDMA induces neurochemical changes in the hippocampus of adolescent and young adult rats: Down-regulation of apoptotic markers. Neurotoxicology 2015; 49:104-13. [DOI: 10.1016/j.neuro.2015.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/05/2015] [Accepted: 06/05/2015] [Indexed: 11/24/2022]
|
33
|
Areal LB, Rodrigues LCM, Andrich F, Moraes LS, Cicilini MA, Mendonça JB, Pelição FS, Nakamura-Palacios EM, Martins-Silva C, Pires RGW. Behavioural, biochemical and molecular changes induced by chronic crack-cocaine inhalation in mice: The role of dopaminergic and endocannabinoid systems in the prefrontal cortex. Behav Brain Res 2015; 290:8-16. [PMID: 25940765 DOI: 10.1016/j.bbr.2015.04.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/16/2015] [Accepted: 04/22/2015] [Indexed: 01/17/2023]
Abstract
Crack-cocaine addiction has increasingly become a public health problem worldwide, especially in developing countries. However, no studies have focused on neurobiological mechanisms underlying the severe addiction produced by this drug, which seems to differ from powder cocaine in many aspects. This study investigated behavioural, biochemical and molecular changes in mice inhaling crack-cocaine, focusing on dopaminergic and endocannabinoid systems in the prefrontal cortex. Mice were submitted to two inhalation sessions of crack-cocaine a day (crack-cocaine group) during 11 days, meanwhile the control group had no access to the drug. We found that the crack-cocaine group exhibited hyperlocomotion and a peculiar jumping behaviour ("escape jumping"). Blood collected right after the last inhalation session revealed that the anhydroecgonine methyl ester (AEME), a specific metabolite of cocaine pyrolysis, was much more concentrated than cocaine itself in the crack-cocaine group. Most genes related to the endocannabinoid system, CB1 receptor and cannabinoid degradation enzymes were downregulated after 11-day crack-cocaine exposition. These changes may have decreased dopamine and its metabolites levels, which in turn may be related with the extreme upregulation of dopamine receptors and tyrosine hydroxylase observed in the prefrontal cortex of these animals. Our data suggest that after 11 days of crack-cocaine exposure, neuroadaptive changes towards downregulation of reinforcing mechanisms may have taken place as a result of neurochemical changes observed on dopaminergic and endocannabinoid systems. Successive changes like these have never been described in cocaine hydrochloride models before, probably because AEME is only produced by cocaine pyrolysis and this metabolite may underlie the more aggressive pattern of addiction induced by crack-cocaine.
Collapse
Affiliation(s)
- Lorena B Areal
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil; Laboratory of Molecular and Behavioral Neurobiology, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil
| | - Livia C M Rodrigues
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil; Laboratory of Cognitive Sciences and Neuropsychopharmacology, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil
| | - Filipe Andrich
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil; Laboratory of Molecular and Behavioral Neurobiology, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil
| | - Livia S Moraes
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil; Laboratory of Molecular and Behavioral Neurobiology, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil
| | - Maria A Cicilini
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil
| | - Josideia B Mendonça
- Laboratory of Forensic Science Service, Espirito Santo State Police, Av. Nossa Senhora. da Penha, 2290, Vitória-ES 29045-402, Brazil
| | - Fabricio S Pelição
- Laboratory of Forensic Science Service, Espirito Santo State Police, Av. Nossa Senhora. da Penha, 2290, Vitória-ES 29045-402, Brazil
| | - Ester M Nakamura-Palacios
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil; Laboratory of Cognitive Sciences and Neuropsychopharmacology, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil
| | - Cristina Martins-Silva
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil; Laboratory of Molecular and Behavioral Neurobiology, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil
| | - Rita G W Pires
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil; Laboratory of Molecular and Behavioral Neurobiology, Health Sciences Center, Federal University of Espirito Santo, Av. Marechal Campos 1468 - Maruípe, Vitoria-ES 29.043-910, Brazil.
| |
Collapse
|
34
|
Sagheddu C, Muntoni AL, Pistis M, Melis M. Endocannabinoid Signaling in Motivation, Reward, and Addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 125:257-302. [DOI: 10.1016/bs.irn.2015.10.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
35
|
Abstract
The physiological and pathophysiological functions of the endocannabinoid system have been studied extensively using transgenic and targeted knockout mouse models. The first gene deletions of the cannabinoid CB(1) receptor were described in the late 1990s, soon followed by CB(2) and FAAH mutations in early 2000. These mouse models helped to elucidate the fundamental role of endocannabinoids as retrograde transmitters in the CNS and in the discovery of many unexpected endocannabinoid functions, for example, in the skin, bone and liver. We now have knockout mouse models for almost every receptor and enzyme of the endocannabinoid system. Conditional mutant mice were mostly developed for the CB(1) receptor, which is widely expressed on many different neurons, astrocytes and microglia, as well as on many cells outside the CNS. These mouse strains include "floxed" CB(1) alleles and mice with a conditional re-expression of CB(1). The availability of these mice made it possible to decipher the function of CB(1) in specific neuronal circuits and cell populations or to discriminate between central and peripheral effects. Many of the genetic mouse models were also used in combination with viral expression systems. The purpose of this review is to provide a comprehensive overview of the existing genetic models and to summarize some of the most important discoveries that were made with these animals.
Collapse
MESH Headings
- Amidohydrolases/genetics
- Amidohydrolases/metabolism
- Animals
- Endocannabinoids/genetics
- Endocannabinoids/metabolism
- Gene Deletion
- Gene Expression Regulation
- Genotype
- Humans
- Hydrolysis
- Mice, Knockout
- Mice, Mutant Strains
- Monoacylglycerol Lipases/genetics
- Monoacylglycerol Lipases/metabolism
- Mutation
- Phenotype
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/genetics
- Receptor, Cannabinoid, CB2/metabolism
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Andreas Zimmer
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany.
| |
Collapse
|
36
|
Abstract
OBJECTIVE Substance dependence disorder is a chronically relapsing condition characterised by neurobiological changes leading to loss of control in restricting a substance intake, compulsion and withdrawal syndrome. In the past few years, (endo)cannabinoids have been raised as a possible target in the aetiology of drug addiction. On the other hand, although the exact mechanisms of the genesis of addiction remain poorly understood, it is possible that neuroinflammation might also play a role in the pathophysiology of this condition. Studies demonstrated that (endo)cannabinoids act as immunomodulators by inhibiting cytokines production and microglial cell activation. Thus, in the present review, we explore the possible role of neuroinflammation on the therapeutic effects of cannabinoids on drug addiction. METHODS We conducted an evidence-based review of the literature in order to assess the role of cannabinoids on the neuroinflammatory hypothesis of addiction (terms: addiction, cannabinoids and inflammation). We searched PubMed and BioMedCentral databases up to April 2014 with no date restrictions. RESULTS In all, 165 eligible articles were included in the present review. Existing evidence suggests that disruption in cannabinoid signalling during the drug addiction process leads to microglial activation and neuroinflammation. CONCLUSION The literature showed that inflammation and changes in endocannabinod signalling occur in drug abuse; however, it remains uncertain whether these changes are causally or coincidentally associated with addiction. Additional studies, therefore, are needed to elucidate the contribution of neuroinflammation on the behavioural and neuroprotective effects of cannabinoids on drug addiction.
Collapse
|
37
|
Franklin JM, Carrasco GA. Cocaine potentiates multiple 5-HT2A receptor signaling pathways and is associated with decreased phosphorylation of 5-HT2A receptors in vivo. J Mol Neurosci 2014; 55:770-7. [PMID: 25213649 DOI: 10.1007/s12031-014-0419-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 08/31/2014] [Indexed: 01/25/2023]
Abstract
Cocaine addiction is a chronic relapsing disorder in which the underlying mechanisms are not well understood. Here, we used Sprague-Dawley rats injected with either saline (1 ml/kg) or cocaine (15 mg/kg) for 7 days (b.i.d, i.p) to study the effect of cocaine on several components of 5-HT2A receptor signaling in prefrontal cortex (PFCx). We detected enhanced activation of 5-HT2A receptor-mediated phospholipase C beta (PLCβ) and extracellular regulated kinase 1/2 activity in PFCx of cocaine-treated rats. Although we were unable to detect changes in the protein levels of several proteins associated with 5-HT2A receptor signaling such as caveolin-1, postsynaptic density protein 95, β-arrestin 2, etc., we found a significant reduction in the phosphorylation status of cortical 5-HT2A receptors. This phenomenon was associated with reduced levels of G-protein receptor kinase 5 (GRK5), but not GRK2 or RSK2, proteins. Our results suggest that decreased phosphorylation of 5-HT2A receptors could mediate, at least in part, the cocaine-induced potentiation of multiple 5-HT2A receptor signaling pathways in rat PFCx. As discussed in this manuscript, we hypothesize that preventing these neuroadaptations in 5-HT2A receptor signaling may alleviate some of the aversive withdrawal-associated symptoms that contribute to relapse to cocaine abuse.
Collapse
Affiliation(s)
- Jade M Franklin
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, 3048B Malott Hall, Lawrence, KS, 66045, USA
| | | |
Collapse
|
38
|
Marchalant Y, Brownjohn PW, Bonnet A, Kleffmann T, Ashton JC. Validating Antibodies to the Cannabinoid CB2 Receptor: Antibody Sensitivity Is Not Evidence of Antibody Specificity. J Histochem Cytochem 2014; 62:395-404. [PMID: 24670796 PMCID: PMC4174627 DOI: 10.1369/0022155414530995] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Antibody-based methods for the detection and quantification of membrane integral proteins, in particular, the G protein-coupled receptors (GPCRs), have been plagued with issues of primary antibody specificity. In this report, we investigate one of the most commonly utilized commercial antibodies for the cannabinoid CB2 receptor, a GPCR, using immunoblotting in combination with mass spectrometry. In this way, we were able to develop powerful negative and novel positive controls. By doing this, we are able to demonstrate that it is possible for an antibody to be sensitive for a protein of interest—in this case CB2—but still cross-react with other proteins and therefore lack specificity. Specifically, we were able to use western blotting combined with mass spectrometry to unequivocally identify CB2 protein in over-expressing cell lines. This shows that a common practice of validating antibodies with positive controls only is insufficient to ensure antibody reliability. In addition, our work is the first to develop a label-free method of protein detection using mass spectrometry that, with further refinement, could provide unequivocal identification of CB2 receptor protein in native tissues.
Collapse
Affiliation(s)
- Yannick Marchalant
- Aix-Marseille University, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie, Marseille, France (YM,AB)Department of Pharmacology and Toxicology (PWB,JCA)Centre for Protein Research, Biochemistry Department, University of Otago, Dunedin, New Zealand (TK)
| | - Philip W Brownjohn
- Aix-Marseille University, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie, Marseille, France (YM,AB)Department of Pharmacology and Toxicology (PWB,JCA)Centre for Protein Research, Biochemistry Department, University of Otago, Dunedin, New Zealand (TK)
| | - Amandine Bonnet
- Aix-Marseille University, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie, Marseille, France (YM,AB)Department of Pharmacology and Toxicology (PWB,JCA)Centre for Protein Research, Biochemistry Department, University of Otago, Dunedin, New Zealand (TK)
| | - Torsten Kleffmann
- Aix-Marseille University, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie, Marseille, France (YM,AB)Department of Pharmacology and Toxicology (PWB,JCA)Centre for Protein Research, Biochemistry Department, University of Otago, Dunedin, New Zealand (TK)
| | - John C Ashton
- Aix-Marseille University, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie, Marseille, France (YM,AB)Department of Pharmacology and Toxicology (PWB,JCA)Centre for Protein Research, Biochemistry Department, University of Otago, Dunedin, New Zealand (TK)
| |
Collapse
|
39
|
Blanco-Calvo E, Rivera P, Arrabal S, Vargas A, Pavón FJ, Serrano A, Castilla-Ortega E, Galeano P, Rubio L, Suárez J, Rodriguez de Fonseca F. Pharmacological blockade of either cannabinoid CB1 or CB2 receptors prevents both cocaine-induced conditioned locomotion and cocaine-induced reduction of cell proliferation in the hippocampus of adult male rat. Front Integr Neurosci 2014; 7:106. [PMID: 24409127 PMCID: PMC3884150 DOI: 10.3389/fnint.2013.00106] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 12/18/2013] [Indexed: 11/14/2022] Open
Abstract
Addiction to major drugs of abuse, such as cocaine, has recently been linked to alterations in adult neurogenesis in the hippocampus. The endogenous cannabinoid system modulates this proliferative response as demonstrated by the finding that pharmacological activation/blockade of cannabinoid CB1 and CB2 receptors not only modulates neurogenesis but also modulates cell death in the brain. In the present study, we evaluated whether the endogenous cannabinoid system affects cocaine-induced alterations in cell proliferation. To this end, we examined whether pharmacological blockade of either CB1 (Rimonabant, 3 mg/kg) or CB2 receptors (AM630, 3 mg/kg) would affect cell proliferation [the cells were labeled with 5-bromo-2′-deoxyuridine (BrdU)] in the subventricular zone (SVZ) of the lateral ventricle and the dentate subgranular zone (SGZ). Additionally, we measured cell apoptosis (as monitored by the expression of cleaved caspase-3) and glial activation [by analyzing the expression of glial fibrillary acidic protein (GFAP) and Iba-1] in the striatum and hippocampus during acute and repeated (4 days) cocaine administration (20 mg/kg). The results showed that acute cocaine exposure decreased the number of BrdU-immunoreactive (ir) cells in the SVZ and SGZ. In contrast, repeated cocaine exposure reduced the number of BrdU-ir cells only in the SVZ. Both acute and repeated cocaine exposure increased the number of cleaved caspase-3-, GFAP- and Iba1-ir cells in the hippocampus, and this effect was counteracted by AM630 or Rimonabant, which increased the number of BrdU-, GFAP-, and Iba1-ir cells in the hippocampus. These results indicate that the changes in neurogenic, apoptotic and gliotic processes that were produced by repeated cocaine administration were normalized by pharmacological blockade of CB1 and CB2. The restorative effects of cannabinoid receptor blockade on hippocampal cell proliferation were associated with the prevention of the induction of conditioned locomotion but not with the prevention of cocaine-induced sensitization.
Collapse
Affiliation(s)
- Eduardo Blanco-Calvo
- Departament de Pedagogia i Psicologia, Facultat de Ciències de l'Educació, Universitat de Lleida Lleida, Spain ; Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Patricia Rivera
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Sergio Arrabal
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Antonio Vargas
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Francisco Javier Pavón
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Antonia Serrano
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Estela Castilla-Ortega
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Pablo Galeano
- Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini, Universidad de Buenos Aires-CONICET Ciudad de Buenos Aires, Argentina
| | - Leticia Rubio
- Departamento de Anatomía y Medicina Legal y Forense, Facultad de Medicina, Universidad de Málaga Málaga, Spain
| | - Juan Suárez
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Fernando Rodriguez de Fonseca
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| |
Collapse
|