1
|
Wei YB, Wang YB, Sun JY, Wang S, Nan J, Yu HL, Lan Y. N-palmitoylethanolamide attenuates negative emotions induced by morphine withdrawal in mice. Neurosci Lett 2024; 841:137944. [PMID: 39154843 DOI: 10.1016/j.neulet.2024.137944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/21/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Depression and anxiety are prominent symptoms of withdrawal syndrome, often caused by the abuse of addictive drugs like morphine. N-palmitoylethanolamide (PEA), a biologically active lipid, is utilized as an anti-inflammatory and analgesic medication. Recent studies have highlighted PEA's role in mitigating cognitive decline and easing depression resulting from chronic pain. However, it remains unknown whether PEA can influence negative emotions triggered by morphine withdrawal. This study seeks to explore the impact of PEA on such emotions and investigate the underlying mechanisms. Mice subjected to morphine treatment underwent a 10-day withdrawal period, followed by assessments of the effect of PEA on anxiety- and depression-like behaviors using various tests. Enzyme-linked immunosorbent assay was conducted to measure levels of monoamine neurotransmitters in specific brain regions. The findings indicate that PEA mitigated anxiety and depression symptoms and reduced 5-hydroxytryptamine, noradrenaline, and dopamine levels in the hippocampus and prefrontal cortex. In summary, PEA demonstrates a significant positive effect on negative emotions associated with morphine withdrawal, accompanied with the reduction in levels of monoamine neurotransmitters in key brain regions. These insights could be valuable for managing negative emotions arising from morphine withdrawal.
Collapse
Affiliation(s)
- Yan-Bin Wei
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji City, Jilin Province 133002, China
| | - Yong-Bo Wang
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji City, Jilin Province 133002, China
| | - Jia-Yue Sun
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji City, Jilin Province 133002, China
| | - Shan Wang
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji City, Jilin Province 133002, China
| | - Jun Nan
- Department of Orthopedics, affiliated Hospital of Yanbian University, Yanji City 133000, China
| | - Hai-Ling Yu
- Department of Functional Science, College of Medicine, Yanbian University, Yanji City, Jilin Province 133002, China
| | - Yan Lan
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji City, Jilin Province 133002, China.
| |
Collapse
|
2
|
Hebert FO, Mongeau-Pérusse V, Rizkallah E, Mahroug A, Bakouni H, Morissette F, Brissette S, Bruneau J, Dubreucq S, Jutras-Aswad D. Absence of Evidence for Sustained Effects of Daily Cannabidiol Administration on Anandamide Plasma Concentration in Individuals with Cocaine Use Disorder: Exploratory Findings from a Randomized Controlled Trial. Cannabis Cannabinoid Res 2024. [PMID: 38770686 DOI: 10.1089/can.2023.0273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
Background: Cannabidiol (CBD) has been proposed to have a therapeutic potential over a wide range of neuropsychiatric disorders, including substance use disorders. Pre-clinical evidence suggests that CBD can increase anandamide (AEA) plasma concentration, possibly mediating some of its therapeutic properties. Whether CBD exerts such an effect on AEA in individuals with cocaine use disorder (CUD) remains unknown. Aims: To explore the sustained effects of daily CBD administration on AEA plasma concentrations compared with placebo in CUD. Methods: We used data from a randomized, double-blind, placebo-controlled trial evaluating CBD's efficacy in CUD. Seventy-eight individuals were randomized to receive a daily oral dose of 800 mg CBD (n = 40) or a placebo (n = 38). Participants stayed in an inpatient detoxification setting for 10 days, after which they were followed in an outpatient setting for 12 weeks. AEA plasma concentration was measured at baseline and at 23-h post CBD ingestion on day 8 and week 4. A generalized estimating equation model was used to assess CBD's effects on AEA, and sensitivity analyses were computed using Bayesian linear regressions. Results: Sixty-four participants were included in the analysis. Similar mean AEA plasma concentrations in both treatment groups (p = 0.357) were observed. At day 8, mean AEA plasma concentrations (± standard deviation) were 0.26 (± 0.07) ng/mL in the CBD group and 0.29 (± 0.08) ng/mL in the placebo group (p = 0.832; Bayes factor [BF] = 0.190). At week 4, they were 0.27 (± 0.09) ng/mL in the CBD group and 0.30 (± 0.09) ng/mL in the placebo group (p = 0.181; BF = 0.194). Conclusion: While not excluding any potential acute and short-term effect, daily CBD administration did not exert a sustained impact on AEA plasma concentrations in individuals with CUD compared with placebo. Registration: clinicaltrials.gov (NCT02559167).
Collapse
Affiliation(s)
| | - Violaine Mongeau-Pérusse
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Elie Rizkallah
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Amani Mahroug
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
| | - Hamzah Bakouni
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Florence Morissette
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Suzanne Brissette
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Family and Emergency Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Julie Bruneau
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Family and Emergency Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Simon Dubreucq
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Didier Jutras-Aswad
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| |
Collapse
|
3
|
Alegre-Zurano L, García-Baos A, Castro-Zavala A, Medrano M, Gallego-Landin I, Valverde O. The FAAH inhibitor URB597 reduces cocaine intake during conditioned punishment and mitigates cocaine seeking during withdrawal. Biomed Pharmacother 2023; 165:115194. [PMID: 37499453 DOI: 10.1016/j.biopha.2023.115194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/30/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
The endocannabinoid system is prominently implicated in the control of cocaine reinforcement due to its relevant role in synaptic plasticity and neurotransmitter modulation in the mesocorticolimbic system. The inhibition of fatty acid amide hydrolase (FAAH), and the resulting increase in anandamide and other N-acylethanolamines, represents a promising strategy for reducing drug seeking. In the present study, we aimed to assess the effects of the FAAH inhibitor URB597 (1 mg/kg) on crucial features of cocaine addictive-like behaviour in mice. Therefore, we tested the effects of URB597 on acquisition of cocaine (0.6 mg/kg/inf) self-administration, compulsive-like cocaine intake and cue-induced drug-seeking behaviour during withdrawal. URB597 reduced cocaine intake under conditioned punishment while having no impact on acquisition. This result was associated to increased cannabinoid receptor 1 gene expression in the ventral striatum and medium spiny neurons activation in the nucleus accumbens shell. Moreover, URB597 mitigated cue-induced drug-seeking behaviour during prolonged abstinence and prevented the withdrawal-induced increase in FAAH gene expression in the ventral striatum. In this case, URB597 decreased activation of medium spiny neurons in the nucleus accumbens core. Our findings evidence the prominent role of endocannabinoids in the development of cocaine addictive-like behaviours and support the potential of FAAH inhibition as a therapeutical target for the treatment of cocaine addiction.
Collapse
Affiliation(s)
- Laia Alegre-Zurano
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Alba García-Baos
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Program, IMIM-Hospital Del Mar Research Institute, Barcelona, Spain
| | - Adriana Castro-Zavala
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Mireia Medrano
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ines Gallego-Landin
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Program, IMIM-Hospital Del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
4
|
Shahen-Zoabi S, Smoum R, Bingor A, Grad E, Nemirovski A, Shekh-Ahmad T, Mechoulam R, Yaka R. N-oleoyl glycine and N-oleoyl alanine attenuate alcohol self-administration and preference in mice. Transl Psychiatry 2023; 13:273. [PMID: 37524707 PMCID: PMC10390512 DOI: 10.1038/s41398-023-02574-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023] Open
Abstract
The endocannabinoid system (ECS) plays a key modulatory role during synaptic plasticity and homeostatic processes in the brain and has an important role in the neurobiological processes underlying drug addiction. We have previously shown that an elevated ECS response to psychostimulant (cocaine) is involved in regulating the development and expression of cocaine-conditioned reward and sensitization. We therefore hypothesized that drug-induced elevation in endocannabinoids (eCBs) and/or eCB-like molecules (eCB-Ls) may represent a protective mechanism against drug insult, and boosting their levels exogenously may strengthen their neuroprotective effects. Here, we determine the involvement of ECS in alcohol addiction. We first measured the eCBs and eCB-Ls levels in different brain reward system regions following chronic alcohol self-administration using LC-MS. We have found that following chronic intermittent alcohol consumption, N-oleoyl glycine (OlGly) levels were significantly elevated in the prefrontal cortex (PFC), and N-oleoyl alanine (OlAla) was significantly elevated in the PFC, nucleus accumbens (NAc) and ventral tegmental area (VTA) in a region-specific manner. We next tested whether exogenous administration of OlGly or OlAla would attenuate alcohol consumption and preference. We found that systemic administration of OlGly or OlAla (60 mg/kg, intraperitoneal) during intermittent alcohol consumption significantly reduced alcohol intake and preference without affecting the hedonic state. These findings suggest that the ECS negatively regulates alcohol consumption and boosting selective eCBs exogenously has beneficial effects against alcohol consumption and potentially in preventing relapse.
Collapse
Affiliation(s)
- Samah Shahen-Zoabi
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Reem Smoum
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Alexey Bingor
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Etty Grad
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Alina Nemirovski
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Tawfeeq Shekh-Ahmad
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Raphael Mechoulam
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Rami Yaka
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
| |
Collapse
|
5
|
González-Portilla M, Mellado S, Montagud-Romero S, Rodríguez de Fonseca F, Pascual M, Rodríguez-Arias M. Oleoylethanolamide attenuates cocaine-primed reinstatement and alters dopaminergic gene expression in the striatum. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2023; 19:8. [PMID: 37226219 DOI: 10.1186/s12993-023-00210-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
The lipid oleoylethanolamide (OEA) has been shown to affect reward-related behavior. However, there is limited experimental evidence about the specific neurotransmission systems OEA may be affecting to exert this modulatory effect. The aim of this study was to evaluate the effects of OEA on the rewarding properties of cocaine and relapse-related gene expression in the striatum and hippocampus. For this purpose, we evaluated male OF1 mice on a cocaine-induced CPP procedure (10 mg/kg) and after the corresponding extinction sessions, we tested drug-induced reinstatement. The effects of OEA (10 mg/kg, i.p.) were evaluated at three different timepoints: (1) Before each cocaine conditioning session (OEA-C), (2) Before extinction sessions (OEA-EXT) and (3) Before the reinstatement test (OEA-REINST). Furthermore, gene expression changes in dopamine receptor D1 gene, dopamine receptor D2 gene, opioid receptor µ, cannabinoid receptor 1, in the striatum and hippocampus were analyzed by qRT-PCR. The results obtained in the study showed that OEA administration did not affect cocaine CPP acquisition. However, mice receiving different OEA treatment schedules (OEA-C, OEA-EXT and OEA-REINST) failed to display drug-induced reinstatement. Interestingly, the administration of OEA blocked the increase of dopamine receptor gene D1 in the striatum and hippocampus caused by cocaine exposure. In addition, OEA-treated mice exhibited reduced striatal dopamine receptor gene D2 and cannabinoid receptor 1. Together, these findings suggest that OEA may be a promising pharmacological agent in the treatment of cocaine use disorder.
Collapse
Affiliation(s)
- Macarena González-Portilla
- Department of Psychobiology, Facultad de Psicología, Universitat de València, Avda. Blasco Ibáñez 21, 46010, Valencia, Spain
| | - Susana Mellado
- Department of Physiology, School of Medicine, Universitat de Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
| | - Sandra Montagud-Romero
- Department of Psychobiology, Facultad de Psicología, Universitat de València, Avda. Blasco Ibáñez 21, 46010, Valencia, Spain
| | - Fernando Rodríguez de Fonseca
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga- IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain
- Atención primaria, cronicidad y promoción de la salud, Red de investigación en atención primaria de adicciones (RIAPAD), Rd210009/0005/0003, Valencia, Madrid, Spain
| | - María Pascual
- Department of Physiology, School of Medicine, Universitat de Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de València, Avda. Blasco Ibáñez 21, 46010, Valencia, Spain.
- Atención primaria, cronicidad y promoción de la salud, Red de investigación en atención primaria de adicciones (RIAPAD), Rd210009/0005/0003, Valencia, Madrid, Spain.
| |
Collapse
|
6
|
Karimi-Haghighi S, Mahmoudi M, Sayehmiri F, Mozafari R, Haghparast A. Endocannabinoid system as a therapeutic target for psychostimulants relapse: A systematic review of preclinical studies. Eur J Pharmacol 2023; 951:175669. [PMID: 36965745 DOI: 10.1016/j.ejphar.2023.175669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/04/2023] [Accepted: 03/20/2023] [Indexed: 03/27/2023]
Abstract
The mechanism behind the reinstament of psychostimulant, as a major obstacle in addiction treatment is not fully understood. Controversial data are available in the literature concerning the role of the endocannabinoid (eCB) system in regulating the relapse to psychostimulant addiction in preclinical studies. The current systematic review aims to evaluate eCB modulators' effect in the reinstatement of commonly abused psychostimulants, including cocaine, amphetamine, methamphetamine, and 3,4-methylenedioxymethamphetamine. By searching the PubMed, Web of Science, and Scopus databases, studies were selected. Then the studies, quality was evaluated by the SYRCLE risk of bias tool. The results have still been limited to preclinical studies. Thirty-nine articles that employed self-administration and CPP as the most prevalent animal models of addiction were selected. This data indicates that cannabinoid receptor 1 antagonists and some cannabinoid receptor 2 agonists could suppress the reinstatement of cocaine and methamphetamine addiction in a dose-dependent manner. However, only AM251 was efficient to block the reinstatement of 3,4-methylenedioxymethamphetamine. In conclusion, cannabinoid receptor 1 antagonists and some cannabinoid receptor 2 agonists may have curative potential in the relapse of psychostimulant abuse. However, time, dose, and route of administration are crucial factors in their inhibitory impacts.
Collapse
Affiliation(s)
- Saeideh Karimi-Haghighi
- Community Based Psychiatric Care Research Center, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maedeh Mahmoudi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sayehmiri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roghayeh Mozafari
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran; Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Lucente E, Söderpalm B, Ericson M, Adermark L. Acute and chronic effects by nicotine on striatal neurotransmission and synaptic plasticity in the female rat brain. Front Mol Neurosci 2023; 15:1104648. [PMID: 36710931 PMCID: PMC9877298 DOI: 10.3389/fnmol.2022.1104648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023] Open
Abstract
Introduction Tobacco use is in part a gendered activity, yet neurobiological studies outlining the effect by nicotine on the female brain are scarce. The aim of this study was to outline acute and sub-chronic effects by nicotine on the female rat brain, with special emphasis on neurotransmission and synaptic plasticity in the dorsolateral striatum (DLS), a key brain region with respect to the formation of habits. Methods In vivo microdialysis and ex vivo electrophysiology were performed in nicotine naïve female Wistar rats, and following sub-chronic nicotine exposure (0.36 mg/kg free base, 15 injections). Locomotor behavior was assessed at the first and last drug-exposure. Results Acute exposure to nicotine ex vivo depresses excitatory neurotransmission by reducing the probability of transmitter release. Bath applied nicotine furthermore facilitated long-term synaptic depression induced by high frequency stimulation (HFS-LTD). The cannabinoid 1 receptor (CB1R) agonist WIN55,212-2 produced a robust synaptic depression of evoked potentials, and HFS-LTD was blocked by the CB1R antagonist AM251, suggesting that HFS-LTD in the female rat DLS is endocannabinoid mediated. Sub-chronic exposure to nicotine in vivo produced behavioral sensitization and electrophysiological recordings performed after 2-8 days abstinence revealed a sustained depression of evoked population spike amplitudes in the DLS, with no concomitant change in paired pulse ratio. Rats receiving sub-chronic nicotine exposure further demonstrated an increased neurophysiological responsiveness to nicotine with respect to both dopaminergic- and glutamatergic signaling. However, a tolerance towards the plasticity facilitating property of bath applied nicotine was developed during sub-chronic nicotine exposure in vivo. In addition, the dopamine D2 receptor agonist quinpirole selectively facilitate HFS-LTD in slices from nicotine naïve rats, suggesting that the tolerance may be associated with changes in dopaminergic signaling. Conclusion Nicotine produces acute and sustained effects on striatal neurotransmission and synaptic plasticity in the female rat brain, which may contribute to the establishment of persistent nicotine taking habits.
Collapse
Affiliation(s)
- Erika Lucente
- Integrative Neuroscience Unit, Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise Adermark
- Integrative Neuroscience Unit, Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,*Correspondence: Louise Adermark, ✉
| |
Collapse
|
8
|
Gawliński D, Gawlińska K, Frankowska M, Filip M. Cocaine and Its Abstinence Condition Modulate Striatal and Hippocampal Wnt Signaling in a Male Rat Model of Drug Self-Administration. Int J Mol Sci 2022; 23:ijms232214011. [PMID: 36430488 PMCID: PMC9693497 DOI: 10.3390/ijms232214011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Recent years have provided more and more evidence confirming the important role of Wnt/β-catenin signaling in the pathophysiology of mental illnesses, including cocaine use disorder. High relapse rates, which is a hallmark of drug addiction, prompt the study of changes in Wnt signaling elements (Wnt5a, Wnt7b, and Ctnnb1) in the motivational aspects of cocaine use and early drug-free period (3 days after the last exposure to cocaine). For this purpose, an animal model of intravenous cocaine self-administration and two types of drug-free period (extinction training and abstinence in the home cage) were used. The studies showed that chronic cocaine self-administration mainly disturbs the expression of Wnt5a and Ctnnb1 (the gene encoding β-catenin) in the examined brain structures (striatum and hippocampus), and the examined types of early abstinence are characterized by a different pattern of changes in the expression of these genes. At the same time, in cocaine self-administrated animals, there were no changes in the level of Wnt5a and β-catenin proteins at the tested time points. Moreover, exposure to cocaine induces a significant reduction in the striatal and hippocampal expression of miR-374 and miR-544, which can regulate Wnt5a levels post-transcriptionally. In summary, previous observations from experimenter-administered cocaine have not been fully validated in the cocaine self-administration model. Yoked cocaine administration appears to disrupt Wnt signaling more than cocaine self-administration. The condition of the cocaine-free period, the routes of drug administration, and the motivational aspect of drug administration play an important role in the type of drug-induced molecular changes observed. Furthermore, in-depth research involving additional brain regions is needed to determine the exact role of Wnt signaling in short-term and long-lasting plasticity as well as in the motivational aspects of cocaine use, and thus to assess its potential as a target for new drug therapy for cocaine use disorder.
Collapse
|
9
|
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: 4.7] [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
|
10
|
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: 2.0] [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
|
11
|
De Sa Nogueira D, Bourdy R, Alcala-Vida R, Filliol D, Andry V, Goumon Y, Zwiller J, Romieu P, Merienne K, Olmstead MC, Befort K. Hippocampal Cannabinoid 1 Receptors Are Modulated Following Cocaine Self-administration in Male Rats. Mol Neurobiol 2022; 59:1896-1911. [PMID: 35032317 DOI: 10.1007/s12035-022-02722-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023]
Abstract
Cocaine addiction is a complex pathology inducing long-term neuroplastic changes that, in turn, contribute to maladaptive behaviors. This behavioral dysregulation is associated with transcriptional reprogramming in brain reward circuitry, although the mechanisms underlying this modulation remain poorly understood. The endogenous cannabinoid system may play a role in this process in that cannabinoid mechanisms modulate drug reward and contribute to cocaine-induced neural adaptations. In this study, we investigated whether cocaine self-administration induces long-term adaptations, including transcriptional modifications and associated epigenetic processes. We first examined endocannabinoid gene expression in reward-related brain regions of the rat following self-administered (0.33 mg/kg intravenous, FR1, 10 days) cocaine injections. Interestingly, we found increased Cnr1 expression in several structures, including prefrontal cortex, nucleus accumbens, dorsal striatum, hippocampus, habenula, amygdala, lateral hypothalamus, ventral tegmental area, and rostromedial tegmental nucleus, with most pronounced effects in the hippocampus. Endocannabinoid levels, measured by mass spectrometry, were also altered in this structure. Chromatin immunoprecipitation followed by qPCR in the hippocampus revealed that two activating histone marks, H3K4Me3 and H3K27Ac, were enriched at specific endocannabinoid genes following cocaine intake. Targeting CB1 receptors using chromosome conformation capture, we highlighted spatial chromatin re-organization in the hippocampus, as well as in the nucleus accumbens, suggesting that destabilization of the chromatin may contribute to neuronal responses to cocaine. Overall, our results highlight a key role for the hippocampus in cocaine-induced plasticity and broaden the understanding of neuronal alterations associated with endocannabinoid signaling. The latter suggests that epigenetic modifications contribute to maladaptive behaviors associated with chronic drug use.
Collapse
Affiliation(s)
- David De Sa Nogueira
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France.,Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Romain Bourdy
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Rafael Alcala-Vida
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Dominique Filliol
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Virginie Andry
- Institut Des Neurosciences Cellulaires Et Intégratives (INCI), UPR 3212, CNRS, 8 Allée du Général Rouvillois, 67000, Strasbourg, France
| | - Yannick Goumon
- Institut Des Neurosciences Cellulaires Et Intégratives (INCI), UPR 3212, CNRS, 8 Allée du Général Rouvillois, 67000, Strasbourg, France
| | - Jean Zwiller
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Pascal Romieu
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Karine Merienne
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Mary C Olmstead
- Department of Psychology, Center for Neuroscience Studies, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Katia Befort
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France.
| |
Collapse
|
12
|
Voegel CD, Kroll SL, Schmid MW, Kexel AK, Baumgartner MR, Kraemer T, Binz TM, Quednow BB. Alterations of Stress-Related Glucocorticoids and Endocannabinoids in Hair of Chronic Cocaine Users. Int J Neuropsychopharmacol 2021; 25:226-237. [PMID: 34676867 PMCID: PMC8929753 DOI: 10.1093/ijnp/pyab070] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/06/2021] [Accepted: 10/20/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Previous research in animals and humans has demonstrated a potential role of stress regulatory systems, such as the hypothalamic-pituitary-adrenal (HPA) axis and the endocannabinoid (eCB) system, in the development of substance use disorders. We thus investigated alterations of HPA and eCB markers in individuals with chronic cocaine use disorder by using an advanced hair analysis technique. METHODS We compared hair concentrations of glucocorticoids (cortisone, cortisol) and the eCBs 2-arachidonylglycerol, anandamide (AEA), oleoylethanolamide (OEA), and palmitoylethanolamide (PEA) between 48 recreational cocaine users (RCU), 25 dependent cocaine users (DCU), and 67 stimulant-naïve controls. Self-reported substance use and hair concentrations of substances were also assessed. RESULTS Significantly higher concentrations of hair cortisone were found in RCU and DCU compared with controls. Hair concentrations of OEA and PEA were significantly lower in DCU compared with RCU and controls. Additionally, within cocaine users, elevated cocaine hair concentration was a significant predictor for increased glucocorticoid and decreased OEA hair levels. Moreover, higher 3,4-methylenedioxymethamphetamine hair concentration was correlated with elevated cortisone and AEA, OEA, and PEA levels in hair within cocaine users, whereas more self-reported cannabis use was associated with lower eCBs levels in hair across the total sample. CONCLUSION Our findings support the hypothesis that the HPA axis and eCB system might be important regulators for substance use disorders. The mechanistic understanding of changes in glucocorticoid and eCB levels in future research might be a promising pharmacological target to reduce stress-induced craving and relapse specifically in cocaine use disorder.
Collapse
Affiliation(s)
- Clarissa D Voegel
- Center for Forensic Hair Analytics, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Sara L Kroll
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy, and Psychosomatics, University of Zurich, Zurich, Switzerland,Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Ann-Kathrin Kexel
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy, and Psychosomatics, University of Zurich, Zurich, Switzerland
| | - Markus R Baumgartner
- Center for Forensic Hair Analytics, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Thomas Kraemer
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Tina M Binz
- Center for Forensic Hair Analytics, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Boris B Quednow
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy, and Psychosomatics, University of Zurich, Zurich, Switzerland,Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology, Zurich, Switzerland,Correspondence: B. B. Quednow, PhD, Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, Zurich, Lenggstrasse 31, CH-8032 Zurich, Switzerland ()
| |
Collapse
|
13
|
Smaga I, Wydra K, Suder A, Sanak M, Caffino L, Fumagalli F, Filip M. Enhancement of the GluN2B subunit of glutamatergic NMDA receptors in rat brain areas after cocaine abstinence. J Psychopharmacol 2021; 35:1226-1239. [PMID: 34587833 DOI: 10.1177/02698811211048283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cocaine use disorder is associated with compulsive drug-seeking and drug-taking, whereas relapse may be induced by several factors, including stress, drug-related places, people, and cues. Recent observations strongly support the involvement of the N-methyl-D-aspartate (NMDA) receptors in cocaine use disorders and abstinence, whereas withdrawal in different environments may affect the intensification of relapse. METHODS The aim of this study was to examine the GluN2B subunit expression and its association with the postsynaptic density protein 95 (PSD95) in several brain structures in rats with a history of cocaine self-administration and housed either in an enriched environment or in an isolated condition. Furthermore, a selective antagonist of the GluN2B subunit-CP 101,606 (10 and 20 mg/kg) administered during exposure to cocaine or a drug-associated conditional stimulus (a cue) was used to evaluate seeking behavior in rats. RESULTS In rats previously self-administering cocaine, we observed an increase in the GluN2B expression in the total homogenate from the dorsal hippocampus under both enriched environment and isolation. Cocaine abstinence under isolation conditions increased the GluN2B and GluN2B/PSD95 complex levels in the PSD fraction of the prelimbic cortex in rats previously self-administering cocaine. Administration of CP 101,606 attenuated cue-induced cocaine-seeking behavior only in isolation-housed rats. CONCLUSION In summary, in this study we showed region-specific changes in both the expression of GluN2B subunit and NMDA receptor trafficking during cocaine abstinence under different housing conditions. Furthermore, we showed that the pharmacological blockade of the GluN2B subunit may be useful in attenuating cocaine-seeking behavior.
Collapse
Affiliation(s)
- Irena Smaga
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Karolina Wydra
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Agata Suder
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Małgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
14
|
Gomes-de-Souza L, Bianchi PC, Costa-Ferreira W, Tomeo RA, Cruz FC, Crestani CC. CB 1 and CB 2 receptors in the bed nucleus of the stria terminalis differently modulate anxiety-like behaviors in rats. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110284. [PMID: 33609604 DOI: 10.1016/j.pnpbp.2021.110284] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 01/01/2023]
Abstract
The endocannabinoid system is implicated in anxiety, but the brain sites involved are not completely understood. The bed nucleus of the stria terminalis (BNST) has been related to anxiety and responses to aversive threats. Besides, endocannabinoid neurotransmission acting via CB1 receptors was identified in the BNST. However, the presence of CB2 receptors and the role of BNST endocannabinoid system in anxiety-like behaviors have never been reported. Therefore, this study investigated the presence of CB1 and CB2 receptors in the BNST and their role in anxiety-like behaviors. For this, gene expression of the endocannabinoid receptors was evaluated in samples from anterior and posterior BNST. Besides, behaviors were evaluated in the elevated plus-maze (EPM) in unstressed rats (trait anxiety-like behavior) and after exposure to restraint stress (restraint-evoked anxiety-like behavior) in rats treated with either the CB1 receptor antagonist AM251 or the CB2 receptor antagonist JTE907 into the anterior BNST. The presence of CB1 and CB2 receptors gene expression was identified in anterior and posterior divisions of the BNST. Bilateral microinjection of AM251 into the anterior BNST dose-dependently increased EPM open arms exploration in unstressed animals and inhibited the anxiety-like behavior in the EPM evoked by restraint. Conversely, intra-BNST microinjection of JTE907 decreased EPM open arms exploration in a dose-dependent manner and inhibited restraint-evoked behavioral changes in the EPM. Taken together, these results indicate that CB1 and CB2 receptors present in the BNST are involved in control of anxiety-like behaviors, and control by the latter is affected by previous stress experience.
Collapse
Affiliation(s)
- Lucas Gomes-de-Souza
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Paula C Bianchi
- Department of Pharmacology, Paulista Medicine School, São Paulo Federal University, São Paulo, Brazil
| | - Willian Costa-Ferreira
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Rodrigo A Tomeo
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Fábio C Cruz
- Department of Pharmacology, Paulista Medicine School, São Paulo Federal University, São Paulo, Brazil
| | - Carlos C Crestani
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil.
| |
Collapse
|
15
|
Pharmacological activation of CB2 receptor protects against ethanol-induced myocardial injury related to RIP1/RIP3/MLKL-mediated necroptosis. Mol Cell Biochem 2020; 474:1-14. [PMID: 32681290 DOI: 10.1007/s11010-020-03828-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/07/2020] [Indexed: 12/16/2022]
Abstract
Chronic ethanol abuse can lead to harmful consequences for the heart, resulting in systolic dysfunction, variability in the heart rate, arrhythmia, and cardiac remodelling. However, the precise molecular mechanism responsible for ethanol-induced cardiomyopathy is poorly understood. In this regard, the present study aimed to describe the RIP1/RIP3/MLKL-mediated necroptotic cell death that may be involved in ethanol-induced cardiomyopathy and characterize CBR-mediated effects on the signalling pathway and myocardial injury. We performed an ethanol vapour administration experiment to analyse the effects of ethanol on cardiac structure and function in male C57BL/6J mice. Ethanol induced a significant decline in the cardiac structure and function, as evidenced by a decline in ejection fraction and fractional shortening, and an increase in serum Creatine Kinase levels, myocardial collagen content, and inflammatory reaction. Furthermore, ethanol also upregulated the expression levels of necroptosis-related markers such as p-RIP1, p-RIP3, and p-MLKL in the myocardium. Nec-1 treatment exerted significant cardioprotective effects by salvaging the heart tissue, improving the cardiac function, and mitigating inflammation and necroptosis. In addition, ethanol abuse caused an imbalance in the endocannabinoid system and regulated two cannabinoid receptors (CB1R and CB2R) in the myocardium. Treatment with selective CB2R agonists, JWH-133 or AM1241, markedly improved the cardiac dysfunction and reduced the ethanol-induced necroptosis in the myocardium. Altogether, our data provide evidence that ethanol abuse-induced cardiotoxicity can possibly be attributed to the RIP1/RIP3/MLKL-mediated necroptosis. Moreover, pharmacological activation of CB2R may represent a new cardioprotective strategy against ethanol-induced cardiotoxicity.
Collapse
|
16
|
Kantak KM. Adolescent-onset vs. adult-onset cocaine use: Impact on cognitive functioning in animal models and opportunities for translation. Pharmacol Biochem Behav 2020; 196:172994. [PMID: 32659242 DOI: 10.1016/j.pbb.2020.172994] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/25/2020] [Accepted: 07/02/2020] [Indexed: 01/03/2023]
Abstract
Animal models are poised to make key contributions to the study of cognitive deficits associated with chronic cocaine use in people. Advantages of animal models include use of a longitudinal experimental design that can control for drug use history and onset-age, sex, drug consumption, and abstinence duration. Twenty-two studies were reviewed (13 in adult male rats, 5 in adolescent vs. adult male rats, 3 in adult male monkeys, and 1 in adult female monkeys), and it was demonstrated repeatedly that male animals with adult-onset cocaine self-administration exposure had impairments in sustained attention, decision making, stimulus-reward learning, working memory, and cognitive flexibility, but not habit learning and spatial learning and memory. These findings have translational relevance because adult cocaine users exhibit a similar range of cognitive deficits. In the limited number of studies available, male rats self-administering cocaine during adolescence were less susceptible than adults to impairment in cognitive flexibility, stimulus-reward learning, and decision making, but were more susceptible than adults to impairment in working memory, a finding also reported in the few studies performed in early-onset cocaine users. These findings suggest that animal models can help fill an unmet need for investigating important but yet-to-be-fully-addressed research questions in people. Research priorities include further investigation of differences between adolescents and adults as well as between males and females following chronic cocaine self-administration. A comprehensive understanding of the broad range of cognitive consequences of chronic cocaine use and the role of developmental plasticity can be of value for improving neuropsychological recovery efforts.
Collapse
Affiliation(s)
- Kathleen M Kantak
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, United States of America.
| |
Collapse
|
17
|
Huggett SB, Stallings MC. Genetic Architecture and Molecular Neuropathology of Human Cocaine Addiction. J Neurosci 2020; 40:5300-5313. [PMID: 32457073 PMCID: PMC7329314 DOI: 10.1523/jneurosci.2879-19.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/04/2020] [Accepted: 05/10/2020] [Indexed: 01/12/2023] Open
Abstract
We integrated genomic and bioinformatic analyses, using data from the largest genome-wide association study of cocaine dependence (CD; n = 6546; 82.37% with CD; 57.39% male) and the largest postmortem gene-expression sample of individuals with cocaine use disorder (CUD; n = 36; 51.35% with CUD; 100% male). Our genome-wide analyses identified one novel gene (NDUFB9) associated with the genetic predisposition to CD in African-Americans. The genetic architecture of CD was similar across ancestries. Individual genes associated with CD demonstrated modest overlap across European-Americans and African-Americans, but the genetic liability for CD converged on many similar tissue types (brain, heart, blood, liver) across ancestries. In a separate sample, we investigated the neuronal gene expression associated with CUD by using RNA sequencing of dorsal-lateral prefrontal cortex neurons. We identified 133 genes differentially expressed between CUD case patients and cocaine-free control subjects, including previously implicated candidates for cocaine use/addiction (FOSB, ARC, KCNJ9/GIRK3, NR4A2, JUNB, and MECP2). Differential expression analyses significantly correlated across European-Americans and African-Americans. While genes significantly associated with CD via genome-wide methods were not differentially expressed, two of these genes (NDUFB9 and C1qL2) were part of a robust gene coexpression network associated with CUD involved in neurotransmission (GABA, acetylcholine, serotonin, and dopamine) and drug addiction. We then used a "guilt-by-association" approach to unravel the biological relevance of NDUFB9 and C1qL2 in the context of CD. In sum, our study furthers the understanding of the genetic architecture and molecular neuropathology of human cocaine addiction and provides a framework for translating biological meaning into otherwise obscure genome-wide associations.SIGNIFICANCE STATEMENT Our study further clarifies the genetic and neurobiological contributions to cocaine addiction, provides a rapid approach for generating testable hypotheses for specific candidates identified by genome-wide research, and investigates the cross-ancestral biological contributions to cocaine use disorder/dependence for individuals of European-American and African-American ancestries.
Collapse
Affiliation(s)
- Spencer B Huggett
- Department of Psychology and Neuroscience, University of Colorado, Boulder, Colorado 80309-0345
- Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado 80309-0447
| | - Michael C Stallings
- Department of Psychology and Neuroscience, University of Colorado, Boulder, Colorado 80309-0345
- Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado 80309-0447
| |
Collapse
|
18
|
Abstract
Research in the cannabinoid field, namely on phytocannabinoids, the endogenous cannabinoids anandamide and 2-arachidonoyl glycerol and their metabolizing and synthetic enzymes, the cannabinoid receptors, and anandamide-like cannabinoid compounds, has expanded tremendously over the last few years. Numerous endocannabinoid-like compounds have been discovered. The Cannabis plant constituent cannabidiol (CBD) was found to exert beneficial effects in many preclinical disease models ranging from epilepsy, cardiovascular disease, inflammation, and autoimmunity to neurodegenerative and kidney diseases and cancer. CBD was recently approved in the United States for the treatment of rare forms of childhood epilepsy. This has triggered the development of many CBD-based products for human use, often with overstated claims regarding their therapeutic effects. In this article, the recently published research on the chemistry and biological effects of plant cannabinoids (specifically CBD), endocannabinoids, certain long-chain fatty acid amides, and the variety of relevant receptors is critically reviewed.
Collapse
Affiliation(s)
- Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20852, USA;
| | - Natalya M Kogan
- Institute for Drug Research, Faculty of Medicine, Hebrew University, Jerusalem 9112102, Israel;
| | - Raphael Mechoulam
- Institute for Drug Research, Faculty of Medicine, Hebrew University, Jerusalem 9112102, Israel;
| |
Collapse
|
19
|
Abstract
Substance use disorder (SUD) is a major public health crisis worldwide, and effective treatment options are limited. During the past 2 decades, researchers have investigated the impact of a variety of pharmacological approaches to treat SUD, one of which is the use of medical cannabis or cannabinoids. Significant progress was made with the discovery of rimonabant, a selective CB1 receptor (CB1R) antagonist (also an inverse agonist), as a promising therapeutic for SUDs and obesity. However, serious adverse effects such as depression and suicidality led to the withdrawal of rimonabant (and almost all other CB1R antagonists/inverse agonists) from clinical trials worldwide in 2008. Since then, much research interest has shifted to other cannabinoid-based strategies, such as peripheral CB1R antagonists/inverse agonists, neutral CB1R antagonists, allosteric CB1R modulators, CB2R agonists, fatty acid amide hydrolase (FAAH) inhibitors, monoacylglycerol lipase (MAGL) inhibitors, fatty acid binding protein (FABP) inhibitors, or nonaddictive phytocannabinoids with CB1R or CB2R-binding profiles, as new therapeutics for SUDs. In this article, we first review recent progress in research regarding the endocannabinoid systems, cannabis reward versus aversion, and the underlying receptor mechanisms. We then review recent progress in cannabinoid-based medication development for the treatment of SUDs. As evidence continues to accumulate, neutral CB1R antagonists (such as AM4113), CB2R agonists (JWH133, Xie2-64), and nonselective phytocannabinoids (cannabidiol, β-caryophyllene, ∆9-tetrahydrocannabivarin) have shown great therapeutic potential for SUDs, as shown in experimental animals. Several cannabinoid-based medications (e.g., dronabinol, nabilone, PF-04457845) that entered clinical trials have shown promising results in reducing withdrawal symptoms in cannabis and opioid users.
Collapse
Affiliation(s)
- Ewa Galaj
- Addiction Biology Unit, Molecular Targets and Medication Discoveries Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Molecular Targets and Medication Discoveries Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA.
| |
Collapse
|