1
|
Moreno-Rodríguez M, Martínez-Gardeazabal J, Bengoetxea de Tena I, Llorente-Ovejero A, Lombardero L, González de San Román E, Giménez-Llort L, Manuel I, Rodríguez-Puertas R. Cognitive improvement via cortical cannabinoid receptors and choline-containing lipids. Br J Pharmacol 2024. [PMID: 39489624 DOI: 10.1111/bph.17381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND AND PURPOSE Recent research linking choline-containing lipids to degeneration of basal forebrain cholinergic neurons in neuropathological states illustrates the challenge of balancing lipid integrity with optimal acetylcholine levels, essential for memory preservation. The endocannabinoid system influences learning and memory processes regulated by cholinergic neurotransmission. Therefore, we hypothesised that activation of the endocannabinoid system may confer neuroprotection against cholinergic degeneration. EXPERIMENTAL APPROACH We examined the neuroprotective potential of sub-chronic treatments with the cannabinoid agonist WIN55,212-2, using ex vivo organotypic tissue cultures including nucleus basalis magnocellularis and cortex and in vivo rat models of specific cholinergic damage induced by 192IgG-saporin. Levels of lipids, choline and acetylcholine were measured with histochemical and immunofluorescence assays, along with [35S]GTPγS autoradiography of cannabinoid and muscarinic GPCRs and MALDI-mass spectrometry imaging analysis. Learning and memory were assessed by the Barnes maze and the novel object recognition test in rats and in the 3xTg-AD mouse model. KEY RESULTS Degeneration, induced by 192IgG-saporin, of baso-cortical cholinergic pathways resulted in memory deficits and decreased cortical levels of lysophosphatidylcholines (LPC). WIN55,212-2 restored cortical cholinergic transmission and LPC levels via activation of cannabinoid receptors. This activation altered cortical lipid homeostasis mainly by reducing sphingomyelins in lesioned animals. These modifications were crucial for memory recovery. CONCLUSION AND IMPLICATIONS We hypothesise that WIN55,212-2 facilitates an alternative choline source by breaking down sphingomyelins, leading to elevated cortical acetylcholine levels and LPCs. These results imply that altering choline-containing lipids via activation of cannabinoid receptors presents a promising therapeutic approach for dementia linked to cholinergic dysfunction.
Collapse
Affiliation(s)
- Marta Moreno-Rodríguez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jonatan Martínez-Gardeazabal
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Iker Bengoetxea de Tena
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Alberto Llorente-Ovejero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Laura Lombardero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | - Lydia Giménez-Llort
- Department of Psychiatry and Forensic Medicine, School of Medicine & Institute of Neuroscience, Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Iván Manuel
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Rafael Rodríguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Barakaldo, Spain
| |
Collapse
|
2
|
Lorenzetti V, Gaillard A, McTavish E, Grace S, Rossetti MG, Batalla A, Bellani M, Brambilla P, Chye Y, Conrod P, Cousijn J, Labuschagne I, Clemente A, Mackey S, Rendell P, Solowij N, Suo C, Li CSR, Terrett G, Thompson PM, Yücel M, Garavan H, Roberts CA. Cannabis Dependence is Associated with Reduced Hippocampal Subregion Volumes Independently of Sex: Findings from an ENIGMA Addiction Working Group Multi-Country Study. Cannabis Cannabinoid Res 2024. [PMID: 38498015 DOI: 10.1089/can.2023.0204] [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: 03/19/2024] Open
Abstract
Background: Males and females who consume cannabis can experience different mental health and cognitive problems. Neuroscientific theories of addiction postulate that dependence is underscored by neuroadaptations, but do not account for the contribution of distinct sexes. Further, there is little evidence for sex differences in the neurobiology of cannabis dependence as most neuroimaging studies have been conducted in largely male samples in which cannabis dependence, as opposed to use, is often not ascertained. Methods: We examined subregional hippocampus and amygdala volumetry in a sample of 206 people recruited from the ENIGMA Addiction Working Group. They included 59 people with cannabis dependence (17 females), 49 cannabis users without cannabis dependence (20 females), and 98 controls (33 females). Results: We found no group-by-sex effect on subregional volumetry. The left hippocampal cornu ammonis subfield 1 (CA1) volumes were lower in dependent cannabis users compared with non-dependent cannabis users (p<0.001, d=0.32) and with controls (p=0.022, d=0.18). Further, the left cornu ammonis subfield 3 (CA3) and left dentate gyrus volumes were lower in dependent versus non-dependent cannabis users but not versus controls (p=0.002, d=0.37, and p=0.002, d=0.31, respectively). All models controlled for age, intelligence quotient (IQ), alcohol and tobacco use, and intracranial volume. Amygdala volumetry was not affected by group or group-by-sex, but was smaller in females than males. Conclusions: Our findings suggest that the relationship between cannabis dependence and subregional volumetry was not moderated by sex. Specifically, dependent (rather than non-dependent) cannabis use may be associated with alterations in selected hippocampus subfields high in cannabinoid type 1 (CB1) receptors and implicated in addictive behavior. As these data are cross-sectional, it is plausible that differences predate cannabis dependence onset and contribute to the initiation of cannabis dependence. Longitudinal neuroimaging work is required to examine the time-course of the onset of subregional hippocampal alterations in cannabis dependence, and their progression as cannabis dependence exacerbates or recovers over time.
Collapse
Affiliation(s)
- Valentina Lorenzetti
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - Alexandra Gaillard
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
- Centre for Mental Health and Department of Health Sciences and Biostatistics, Swinburne University, Hawthorn, Australia
| | - Eugene McTavish
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - Sally Grace
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - Maria Gloria Rossetti
- UOC Psichiatria, Azienda Ospedaliera Universitaria Integrata (AOUI), Verona, Italy
- Section of Psychiatry, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Verona, Italy
| | - Albert Batalla
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marcella Bellani
- Section of Psychiatry, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Verona, Italy
| | - Paolo Brambilla
- UOC Psichiatria, Azienda Ospedaliera Universitaria Integrata (AOUI), Verona, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Yann Chye
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Patricia Conrod
- Department of Psychiatry, Université de Montreal, CHU Ste Justine Hospital, Montreal, Canada
| | - Janna Cousijn
- Neuroscience of Addiction Lab, Center for Substance Use and Addiction Research (CESAR), Department of Psychology, Education & Child Studies, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Izelle Labuschagne
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
- School of Psychology, Faculty of Health and Behavioural Sciences, University of Queensland, St Lucia, Australia
| | - Adam Clemente
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - Scott Mackey
- Department of Psychiatry, University of Vermont, Burlington, Vermont, USA
| | - Peter Rendell
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
- School of Psychology, Faculty of Health and Behavioural Sciences, University of Queensland, St Lucia, Australia
| | - Nadia Solowij
- School of Psychology, Faculty of the Arts, Social Sciences and Humanities, University of Wollongong, Wollongong, Australia
| | - Chao Suo
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gill Terrett
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - Paul M Thompson
- Department of Neurology, Imaging Genetics Center, Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Murat Yücel
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Hugh Garavan
- School of Psychology, Faculty of Health and Behavioural Sciences, University of Queensland, St Lucia, Australia
| | - Carl A Roberts
- Department of Psychology, Institute of Population Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
3
|
Fartootzadeh R, Taheri RA, Nourani MR, Goudarzi K. Orexin and cannabinoid systems modulate long-term potentiation of the hippocampus CA1 area in anesthetized rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:461-465. [PMID: 38419882 PMCID: PMC10897562 DOI: 10.22038/ijbms.2023.73979.16075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/20/2023] [Indexed: 03/02/2024]
Abstract
Objectives Long-term potentiation (LTP) is a kind of synaptic plasticity and has a key role in learning and memory. Endocannabinoids and orexins are the endogenous systems that can modulate synaptic plasticity. Given that new studies have shown an interaction between cannabinoid and orexin systems in the brain, we decided to examine this interaction between the two systems on LTP induction in rat's hippocampus. Materials and Methods Twenty-eight male Wistar rats were used for evaluating the effects of co-administrating of cannabinoid-1 receptor (CB1R) antagonist (AM251) and orexin-2 receptor (OX2R) antagonist (TCS OX2 29) on the induction of LTP in the Schaffer collateral-CA1 synapses of rat hippocampus. The drugs were microinjected into the CA1 area of rat hippocampus 30 min before inducing of LTP. Results Results showed that sole administration of the antagonists inhibited LTP, with respect to the control group. Also, co-administrating of them reduced LTP as compared to the control group, but not significantly more than that when the antagonists were solely microinjected into the CA1. Nonetheless, the inhibitory effect of concurrent administration of the antagonists on LTP lasted until the end of the recording. Conclusion These results propose that endogenous cannabinoids and orexins play a role in the expression of LTP, at least by CA1-CB1Rs and CA1-OX2Rs, respectively. Finally, there is no interaction between CB1R and OX2R on the induction of LTP in the Schaffer collateral-CA1 synapses; therefore, these two systems possibly act through common signaling pathways in the hippocampus's CA1 region.
Collapse
Affiliation(s)
- Reza Fartootzadeh
- Department of Physiology, Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
- Department of Physiology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ramezan Ali Taheri
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Nourani
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Kamelya Goudarzi
- Department of Psychiatry, Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| |
Collapse
|
4
|
Rezayof A, Ghasemzadeh Z, Sahafi OH. Addictive drugs modify neurogenesis, synaptogenesis and synaptic plasticity to impair memory formation through neurotransmitter imbalances and signaling dysfunction. Neurochem Int 2023; 169:105572. [PMID: 37423274 DOI: 10.1016/j.neuint.2023.105572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 07/11/2023]
Abstract
Drug abuse changes neurophysiological functions at multiple cellular and molecular levels in the addicted brain. Well-supported scientific evidence suggests that drugs negatively affect memory formation, decision-making and inhibition, and emotional and cognitive behaviors. The mesocorticolimbic brain regions are involved in reward-related learning and habitual drug-seeking/taking behaviors to develop physiological and psychological dependence on the drugs. This review highlights the importance of specific drug-induced chemical imbalances resulting in memory impairment through various neurotransmitter receptor-mediated signaling pathways. The mesocorticolimbic modifications in the expression levels of brain-derived neurotrophic factor (BDNF) and the cAMP-response element binding protein (CREB) impair reward-related memory formation following drug abuse. The contributions of protein kinases and microRNAs (miRNAs), along with the transcriptional and epigenetic regulation have also been considered in memory impairment underlying drug addiction. Overall, we integrate the research on various types of drug-induced memory impairment in distinguished brain regions and provide a comprehensive review with clinical implications addressing the upcoming studies.
Collapse
Affiliation(s)
- Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Zahra Ghasemzadeh
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Oveis Hosseinzadeh Sahafi
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
5
|
Chen C. Inhibiting degradation of 2-arachidonoylglycerol as a therapeutic strategy for neurodegenerative diseases. Pharmacol Ther 2023; 244:108394. [PMID: 36966972 PMCID: PMC10123871 DOI: 10.1016/j.pharmthera.2023.108394] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Endocannabinoids are endogenous lipid signaling mediators that participate in a variety of physiological and pathological processes. 2-Arachidonoylglycerol (2-AG) is the most abundant endocannabinoid and is a full agonist of G-protein-coupled cannabinoid receptors (CB1R and CB2R), which are targets of Δ9-tetrahydrocannabinol (Δ9-THC), the main psychoactive ingredient in cannabis. While 2-AG has been well recognized as a retrograde messenger modulating synaptic transmission and plasticity at both inhibitory GABAergic and excitatory glutamatergic synapses in the brain, growing evidence suggests that 2-AG also functions as an endogenous terminator of neuroinflammation in response to harmful insults, thus maintaining brain homeostasis. Monoacylglycerol lipase (MAGL) is the key enzyme that degrades 2-AG in the brain. The immediate metabolite of 2-AG is arachidonic acid (AA), a precursor of prostaglandins (PGs) and leukotrienes. Several lines of evidence indicate that pharmacological or genetic inactivation of MAGL, which boosts 2-AG levels and reduces its hydrolytic metabolites, resolves neuroinflammation, mitigates neuropathology, and improves synaptic and cognitive functions in animal models of neurodegenerative diseases, including Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and traumatic brain injury (TBI)-induced neurodegenerative disease. Thus, it has been proposed that MAGL is a potential therapeutic target for treatment of neurodegenerative diseases. As the main enzyme hydrolyzing 2-AG, several MAGL inhibitors have been identified and developed. However, our understanding of the mechanisms by which inactivation of MAGL produces neuroprotective effects in neurodegenerative diseases remains limited. A recent finding that inhibition of 2-AG metabolism in astrocytes, but not in neurons, protects the brain from TBI-induced neuropathology might shed some light on this unsolved issue. This review provides an overview of MAGL as a potential therapeutic target for neurodegenerative diseases and discusses possible mechanisms underlying the neuroprotective effects of restraining degradation of 2-AG in the brain.
Collapse
|
6
|
Niloy N, Hediyal TA, Vichitra C, Sonali S, Chidambaram SB, Gorantla VR, Mahalakshmi AM. Effect of Cannabis on Memory Consolidation, Learning and Retrieval and Its Current Legal Status in India: A Review. Biomolecules 2023; 13:biom13010162. [PMID: 36671547 PMCID: PMC9855787 DOI: 10.3390/biom13010162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Cannabis is one of the oldest crops grown, traditionally held religious attachments in various cultures for its medicinal use much before its introduction to Western medicine. Multiple preclinical and clinical investigations have explored the beneficial effects of cannabis in various neurocognitive and neurodegenerative diseases affecting the cognitive domains. Tetrahydrocannabinol (THC), the major psychoactive component, is responsible for cognition-related deficits, while cannabidiol (CBD), a non-psychoactive phytocannabinoid, has been shown to elicit neuroprotective activity. In the present integrative review, the authors focus on the effects of cannabis on the different cognitive domains, including learning, consolidation, and retrieval. The present study is the first attempt in which significant focus has been imparted on all three aspects of cognition, thus linking to its usage. Furthermore, the investigators have also depicted the current legal position of cannabis in India and the requirement for reforms.
Collapse
Affiliation(s)
- Nandi Niloy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Tousif Ahmed Hediyal
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Chandrasekaran Vichitra
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Sharma Sonali
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Vasavi Rakesh Gorantla
- Department of Anatomical Science, St. George’s University, University Centre, St. Georges FZ818, Grenada
- Correspondence: (V.R.G.); (A.M.M.)
| | - Arehally M. Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Correspondence: (V.R.G.); (A.M.M.)
| |
Collapse
|
7
|
Nazari M, Karimi SA, Komaki S, Kourosh Arami M, Komaki A. Underlying mechanisms of long-term potentiation during the inhibition of the cannabinoid CB1 and GABAB receptors in the dentate gyrus of hippocampus. BMC Neurosci 2023; 24:3. [PMID: 36635629 PMCID: PMC9835329 DOI: 10.1186/s12868-022-00767-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/13/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The release of various neurotransmitters and thereby the excitability of neuronal circuits are regulated by the endocannabinoid system in an activity-dependent manner. Hippocampal long-term potentiation (LTP) is augmented in cannabinoid type 1 (CB1) receptor-deficient mice. CB1 receptors exist on GABAergic axon terminals in the hippocampus. In our previous work, we showed that CB1 antagonists increased the population spike (PS) amplitude, field excitatory post-synaptic potential (fEPSP), and the LTP induction in the dentate gyrus (DG) of the rat hippocampus while the GABAB antagonist decreased these parameters. Determining the underlying mechanisms of the pre- and/or postsynaptic locus of LTP expression is of great importance. In this study, we investigated whether LTP alteration acutely caused by CB1 and GABAB receptor antagonists (AM251 and CGP55845, respectively) happens at the postsynaptic or presynaptic regions, or at both. Therefore, the paired-pulse ratio (PPR) was assessed prior to and following the LTP induction in the studied groups. METHODS Male Wistar rats were randomly assigned to the groups of control, AM251, CGP55845, CGP55845 + AM251. A high-frequency stimulation (HFS) of the perforant path (PP) was used to induce LTP in the DG region. RESULTS Statistical analysis revealed that AM251 produced significant increase in excitatory postsynaptic potential (EPSP) slope and amplitude of PS. Conversely, administration of CGP55845 produced decrease in slope of EPSP. The current results indicated that the PPR was not influenced by LTP induction in the presence of AM251 or CGP55845 either alone or their combination. CONCLUSIONS It can be concluded that the site causing LTP expression is, at least in part, the postsynaptic site because PPR was not influenced by LTP induction in the presence of AM251 or CGP55845 either alone or their combination.
Collapse
Affiliation(s)
- Masoumeh Nazari
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, 65178/518, Hamadan, Iran
| | - Seyed Asaad Karimi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, 65178/518, Hamadan, Iran
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Somayeh Komaki
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, 65178/518, Hamadan, Iran
| | - Masoumeh Kourosh Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Komaki
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, 65178/518, Hamadan, Iran.
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
8
|
Ostlund I, Von Gunten M, Smith C, Edwards JG. Chronic Δ9-tetrahydrocannabinol impact on plasticity, and differential activation requirement for CB1-dependent long-term depression in ventral tegmental area GABA neurons in adult versus young mice. Front Neurosci 2023; 16:1067493. [PMID: 36699526 PMCID: PMC9869137 DOI: 10.3389/fnins.2022.1067493] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/12/2022] [Indexed: 01/11/2023] Open
Abstract
The ventral tegmental area (VTA) mediates incentive salience and reward prediction error through dopamine (DA) neurons that are regulated by local VTA GABA neurons. In young mice, VTA GABA cells exhibit a form of synaptic plasticity known as long-term depression (LTD) that is dependent on cannabinoid 1 (CB1) receptors preceded by metabotropic glutamate receptor 5 (mGluR5) signaling to induce endocannabinoid production. This LTD was eliminated following chronic (7-10 consecutive days) exposure to the marijuana derived cannabinoid Δ9 -tetrahydrocannabinol (THC). We now examine the mechanism behind THC-induced elimination of LTD in adolescents as well as plasticity induction ability in adult versus young male and female mice using whole-cell electrophysiology experiments of VTA GABA cells. Chronic THC injections in adolescents resulted in a loss of CB1 agonist-mediated depression, illustrating chronic THC likely desensitizes or removes synaptic CB1. We noted that seven days withdrawal from chronic THC restored LTD and CB1 agonist-induced depression, suggesting reversibility of THC-induced changes. Adult mice continue to express functional mGluR5 and CB1, but require a doubling of the synaptic stimulation compared to young mice to induce LTD, suggesting a quantitative difference in CB1-dependent plasticity between young and adult mice. One potential rationale for this difference is changes in AMPA and NMDA glutamate receptors. Indeed, AMPA/NMDA ratios were increased in in adults compared to young mice. Lastly, we performed quantitative reverse-transcription PCR and identified that CB1, DAGLα, and GluA1 levels increased following chronic THC exposure. Collectively, our data demonstrate the first age-dependent GABA neuron plasticity in the VTA, which could have implications for decreased THC dependence capacity in adults, as well as the mechanism behind chronic THC-induced synaptic alterations in young mice.
Collapse
Affiliation(s)
- Isaac Ostlund
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
| | | | - Calvin Smith
- Neuroscience Center, Brigham Young University, Provo, UT, United States
| | - Jeffrey G. Edwards
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
- Neuroscience Center, Brigham Young University, Provo, UT, United States
| |
Collapse
|
9
|
Lemtiri-Chlieh F, Levine ES. 2-AG and anandamide enhance hippocampal long-term potentiation via suppression of inhibition. Front Cell Neurosci 2022; 16:1023541. [PMID: 36212685 PMCID: PMC9534525 DOI: 10.3389/fncel.2022.1023541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
It is widely accepted that exogenous cannabinoids can impair short-term memory and cognition in humans and other animals. This is likely related to the inhibition of long-term potentiation (LTP), a form of synaptic plasticity, by the global and sustained activation of CB1 cannabinoid receptors in the presence of exogenous agonists. Conversely, the temporally and spatially restricted release of endogenous cannabinoid (eCB) ligands may enhance synaptic plasticity in a synapse-specific manner. We examined the role of eCB signaling in LTP by recording field excitatory postsynaptic potentials (fEPSPs) in the CA1 stratum radiatum in hippocampal slices from juvenile mice. LTP was induced either electrically, by theta burst stimulation (TBS), or pharmacologically, by treatment for 15 min with a solution designed to increase intracellular cAMP (chem-LTP). A stable and long-lasting potentiation in fEPSP slope following TBS was significantly reduced by blocking cannabinoid receptor activation with CB1 receptor antagonists. Chem-LTP caused a sustained 2-fold increase in fEPSP slope and was also blocked by CB1 receptor antagonists. TBS-LTP was partially reduced by inhibiting the synthesis of the endogenous ligands 2-arachidonylglycerol (2-AG) and anandamide. A similar effect was observed with chem-LTP. Blocking inhibitory synapses completely prevented the effect of CB1 receptor antagonists or inhibition of eCB synthesis on TBS-LTP and chem-LTP. These results indicate that simultaneous activation of CB1 receptors by 2-AG and anandamide enhances TBS-induced and pharmacologically-induced LTP, and this effect is mediated by the suppression of inhibition at GABAergic synapses.
Collapse
|
10
|
Augustin SM, Lovinger DM. Synaptic changes induced by cannabinoid drugs and cannabis use disorder. Neurobiol Dis 2022; 167:105670. [DOI: 10.1016/j.nbd.2022.105670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/10/2022] [Accepted: 02/21/2022] [Indexed: 10/19/2022] Open
|
11
|
Ballestar-Tarín ML, Ibáñez-del-Valle V, Cauli O, Navarro-Martínez R. Personal and Social Consequences of Psychotropic Substance Use: A Population-Based Internet Survey. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:65. [PMID: 35056373 PMCID: PMC8777796 DOI: 10.3390/medicina58010065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 11/17/2022]
Abstract
Background and objectives: Drug abuse has become a major worldwide health concern among all age groups. The present study analyses substance misuse and its social and personal consequences using a population-based internet survey in Spain. Materials and Methods: Screening for drug abuse (of alcohol, marijuana/hashish and psychostimulants) and its related risks and problems was performed using the Car, Relax, Alone, Forget, Friends, Trouble (CRAFFT) score. Socio-demographic factors, depressive, anxiety and stress symptoms as well as health habits were also evaluated. We used Linear regression methods to compare each variable's individual contribution so as to determine which one best explains the results. Results: In this population-based study, 1224 people completed and returned the online survey. Of all participants, 57% reported consuming at least one substance based on the CRAFFT scale. While increasing age reduces the probability of personal and social consequences of consumption, people who smoke receive up to three times more (OR = 3.370) recommendations from family and friends to reduce their consumption. As for the type of substance, the consumption of marijuana increases the risk of forgetting (OR = 2.33) and the consumption of other psychostimulant substances almost triples the risk of consuming alone (OR = 2.965). Combining substances can increase the rate of driving a vehicle after consumption by 3.4 times. Conclusions: Although age, smoking and the type of substances used increase the risk of suffering from social and personal consequences of the use or abuse of substances, future studies are needed to determine the influence of new variables as a potential tool for treating and minimizing the adverse consequences of drug abuse.
Collapse
Affiliation(s)
- María Luisa Ballestar-Tarín
- Department of Nursing, Faculty of Nursing and Podiatry, University of Valencia, Avda Menéndez Pidal 19, 46010 Valencia, Spain; (M.L.B.-T.); (V.I.-d.-V.); (R.N.-M.)
| | - Vanessa Ibáñez-del-Valle
- Department of Nursing, Faculty of Nursing and Podiatry, University of Valencia, Avda Menéndez Pidal 19, 46010 Valencia, Spain; (M.L.B.-T.); (V.I.-d.-V.); (R.N.-M.)
- Frailty and Cognitive Impairment Organized Group (FROG), Department of Nursing, University of Valencia, Avda Menéndez Pidal 19, 46010 Valencia, Spain
| | - Omar Cauli
- Department of Nursing, Faculty of Nursing and Podiatry, University of Valencia, Avda Menéndez Pidal 19, 46010 Valencia, Spain; (M.L.B.-T.); (V.I.-d.-V.); (R.N.-M.)
- Frailty and Cognitive Impairment Organized Group (FROG), Department of Nursing, University of Valencia, Avda Menéndez Pidal 19, 46010 Valencia, Spain
| | - Rut Navarro-Martínez
- Department of Nursing, Faculty of Nursing and Podiatry, University of Valencia, Avda Menéndez Pidal 19, 46010 Valencia, Spain; (M.L.B.-T.); (V.I.-d.-V.); (R.N.-M.)
- Frailty and Cognitive Impairment Organized Group (FROG), Department of Nursing, University of Valencia, Avda Menéndez Pidal 19, 46010 Valencia, Spain
| |
Collapse
|
12
|
Marszalek-Grabska M, Smaga I, Surowka P, Grochecki P, Slowik T, Filip M, Kotlinska JH. Memantine Prevents the WIN 55,212-2 Evoked Cross-Priming of Ethanol-Induced Conditioned Place Preference (CPP). Int J Mol Sci 2021; 22:ijms22157940. [PMID: 34360704 PMCID: PMC8348856 DOI: 10.3390/ijms22157940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 11/16/2022] Open
Abstract
The activation of the endocannabinoid system controls the release of many neurotransmitters involved in the brain reward pathways, including glutamate. Both endocannabinoid and glutamate systems are crucial for alcohol relapse. In the present study, we hypothesize that N-methyl-D-aspartate (NMDA) glutamate receptors regulate the ability of a priming dose of WIN 55,212-2 to cross-reinstate ethanol-induced conditioned place preference (CPP). To test this hypothesis, ethanol-induced (1.0 g/kg, 10% w/v, i.p.) CPP (unbiased method) was established using male adult Wistar rats. After CPP extinction, one group of animals received WIN 55,212-2 (1.0 and 2.0 mg/kg, i.p.), the cannabinoid receptor 1 (CB1) agonist, or ethanol, and the other group received memantine (3.0 or 10 mg/kg, i.p.), the NMDA antagonist and WIN 55,212-2 on the reinstatement day. Our results showed that a priming injection of WIN 55,212-2 (2.0 mg/kg, i.p.) reinstated (cross-reinstated) ethanol-induced CPP with similar efficacy to ethanol. Memantine (3.0 or 10 mg/kg, i.p.) pretreatment blocked this WIN 55,212-2 effect. Furthermore, our experiments indicated that ethanol withdrawal (7 days withdrawal after 10 days ethanol administration) down-regulated the CNR1 (encoding CB1), GRIN1/2A (encoding GluN1 and GluN2A subunit of the NMDA receptor) genes expression in the prefrontal cortex and dorsal striatum, but up-regulated these in the hippocampus, confirming the involvement of these receptors in ethanol rewarding effects. Thus, our results show that the endocannabinoid system is involved in the motivational properties of ethanol, and glutamate may control cannabinoid induced relapse into ethanol seeking behavior.
Collapse
Affiliation(s)
- Marta Marszalek-Grabska
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Irena Smaga
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (I.S.); (M.F.)
| | - Paulina Surowka
- Affective Cognitive Neuroscience Laboratory, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland;
| | - Pawel Grochecki
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Tymoteusz Slowik
- Experimental Medicine Center, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Malgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (I.S.); (M.F.)
| | - Jolanta H. Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 20-093 Lublin, Poland;
- Correspondence:
| |
Collapse
|
13
|
Irrera N, Bitto A, Sant’Antonio E, Lauro R, Musolino C, Allegra A. Pros and Cons of the Cannabinoid System in Cancer: Focus on Hematological Malignancies. Molecules 2021; 26:molecules26133866. [PMID: 34202812 PMCID: PMC8270322 DOI: 10.3390/molecules26133866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/09/2021] [Accepted: 06/20/2021] [Indexed: 11/23/2022] Open
Abstract
The endocannabinoid system (ECS) is a composite cell-signaling system that allows endogenous cannabinoid ligands to control cell functions through the interaction with cannabinoid receptors. Modifications of the ECS might contribute to the pathogenesis of different diseases, including cancers. However, the use of these compounds as antitumor agents remains debatable. Pre-clinical experimental studies have shown that cannabinoids (CBs) might be effective for the treatment of hematological malignancies, such as leukemia and lymphoma. Specifically, CBs may activate programmed cell death mechanisms, thus blocking cancer cell growth, and may modulate both autophagy and angiogenesis. Therefore, CBs may have significant anti-tumor effects in hematologic diseases and may synergistically act with chemotherapeutic agents, possibly also reducing chemoresistance. Moreover, targeting ECS might be considered as a novel approach for the management of graft versus host disease, thus reducing some symptoms such as anorexia, cachexia, fatigue, anxiety, depression, and neuropathic pain. The aim of the present review is to collect the state of the art of CBs effects on hematological tumors, thus focusing on the essential topics that might be useful before moving into the clinical practice.
Collapse
Affiliation(s)
- Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (N.I.); (A.B.); (R.L.)
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (N.I.); (A.B.); (R.L.)
| | | | - Rita Lauro
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (N.I.); (A.B.); (R.L.)
| | - Caterina Musolino
- Department of Human Pathology in Adulthood and Childhood, University of Messina, 98125 Messina, Italy;
| | - Alessandro Allegra
- Department of Human Pathology in Adulthood and Childhood, University of Messina, 98125 Messina, Italy;
- Correspondence: ; Tel.: +390902212364
| |
Collapse
|
14
|
Chahkandi M, Komeili G, Sepehri G, Khaksari M, Amiresmaili S. Marijuana and β-estradiol interactions on spatial learning and memory in young female rats: Lack of role of the G protein-coupled estrogen receptor (GPR30). Life Sci 2021; 280:119723. [PMID: 34146552 DOI: 10.1016/j.lfs.2021.119723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 11/18/2022]
Abstract
It has been shown that 17β-estradiol (E2) hormone is an essential biological factor for increasing the sensitivity of women to drug abuse. Recent studies have shown a potential overlap between the molecular pathways of cannabinoids and ovarian hormones. The current study evaluated the interference between the marijuana and E2 effect on spatial learning and memory and the role of the G protein-coupled estrogen receptor (GPR30) in young female rats. The animals were separated into two main groups: intact-ovary and ovariectomized (OVX) rats. The latter group received intraperitoneal injections of E2, G-1 (GPR30 agonist), G-15 (GPR30 antagonist), marijuana, and different combinations of these substances for 28 days. Spatial learning and memory were evaluated by the Morris water maze (MWM) test. We also assessed the BDNF (brain-derived neurotrophic factor) concentration and the hippocampal level of GPR30. The results showed a significant reduction of spatial learning and memory in OVX rats compared to intact-ovary rats, which were restored by E2 replacement. Moreover, treatment with G-1 mimicked E2 effects on spatial learning and memory. Marijuana impaired spatial learning and memory in intact-ovary rats, while improved in OVX rats. We also found that treatment with M + E2 induced significant impairment in spatial learning and memory; however, treatment with M + G1 and M + G15 + E2 showed no significant difference. No significant differences in BDNF expression were observed in experimental groups. These results suggest that marijuana and E2 interact in their effect on spatial learning and memory in young female rats, but GPR30 seems to play no role in this interaction.
Collapse
Affiliation(s)
- Mohadeseh Chahkandi
- Physiology Research Center, Institute of Neuropharmacology, and Department of Physiology and Pharmacology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Komeili
- Department of Physiology, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Gholamreza Sepehri
- Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
15
|
Kesner AJ, Lovinger DM. Cannabis use, abuse, and withdrawal: Cannabinergic mechanisms, clinical, and preclinical findings. J Neurochem 2021; 157:1674-1696. [PMID: 33891706 PMCID: PMC9291571 DOI: 10.1111/jnc.15369] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022]
Abstract
Cannabis sativa is the most widely used illicit drug in the world. Its main psychoactive component is delta-9-tetrahydrocannabinol (THC), one of over 100 phytocannabinoid compounds produced by the cannabis plant. THC is the primary compound that drives cannabis abuse potential and is also used and prescribed medically for therapeutic qualities. Despite its therapeutic potential, a significant subpopulation of frequent cannabis or THC users will develop a drug use syndrome termed cannabis use disorder. Individuals suffering from cannabis use disorder exhibit many of the hallmarks of classical addictions including cravings, tolerance, and withdrawal symptoms. Currently, there are no efficacious treatments for cannabis use disorder or withdrawal symptoms. This makes both clinical and preclinical research on the neurobiological mechanisms of these syndromes ever more pertinent. Indeed, basic research using animal models has provided valuable evidence of the neural molecular and cellular actions of cannabis that mediate its behavioral effects. One of the main components being central action on the cannabinoid type-one receptor and downstream intracellular signaling related to the endogenous cannabinoid system. Back-translational studies have provided insight linking preclinical basic and behavioral biology research to better understand symptoms observed at the clinical level. This narrative review aims to summarize major research elucidating the molecular, cellular, and behavioral manifestations of cannabis/THC use that play a role in cannabis use disorder and withdrawal.
Collapse
Affiliation(s)
- Andrew J. Kesner
- Laboratory for Integrative NeuroscienceNational Institute on Alcohol Abuse and AlcoholismCenter on Compulsive BehaviorsNational Institutes of HealthBethesdaMDUSA
| | - David M. Lovinger
- Laboratory for Integrative NeuroscienceNational Institute on Alcohol Abuse and AlcoholismCenter on Compulsive BehaviorsNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
16
|
Hoffman AF, Hwang EK, Lupica CR. Impairment of Synaptic Plasticity by Cannabis, Δ 9-THC, and Synthetic Cannabinoids. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a039743. [PMID: 32341064 PMCID: PMC8091957 DOI: 10.1101/cshperspect.a039743] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The ability of neurons to dynamically and flexibly encode synaptic inputs via short- and long-term plasticity is critical to an organism's ability to learn and adapt to the environment. Whereas synaptic plasticity may be encoded by pre- or postsynaptic mechanisms, current evidence suggests that optimization of learning requires both forms of plasticity. Endogenous cannabinoids (eCBs) play critical roles in modulating synaptic transmission via activation of cannabinoid CB1 receptors (CB1Rs) in many central nervous system (CNS) regions, and the eCB system has been implicated, either directly or indirectly, in several forms of synaptic plasticity. Because of this, perturbations within the eCB signaling system can lead to impairments in a variety of learned behaviors. One agent of altered eCB signaling is exposure to "exogenous cannabinoids" such as the primary psychoactive constituent of cannabis, Δ9-THC, or illicit synthetic cannabinoids that in many cases have higher potency and efficacy than Δ9-THC. Thus, by targeting the eCB system, these agonists can produce widespread impairment of synaptic plasticity by disrupting ongoing eCB function. Here, we review studies in which Δ9-THC and synthetic cannabinoids impair synaptic plasticity in a variety of neuronal circuits and examine evidence that this contributes to their well-documented ability to disrupt cognition and behavior.
Collapse
Affiliation(s)
- Alexander F Hoffman
- Electrophysiology Research Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Eun-Kyung Hwang
- Electrophysiology Research Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Carl R Lupica
- Electrophysiology Research Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| |
Collapse
|
17
|
Avchalumov Y, Mandyam CD. Plasticity in the Hippocampus, Neurogenesis and Drugs of Abuse. Brain Sci 2021; 11:404. [PMID: 33810204 PMCID: PMC8004884 DOI: 10.3390/brainsci11030404] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/05/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023] Open
Abstract
Synaptic plasticity in the hippocampus assists with consolidation and storage of long-lasting memories. Decades of research has provided substantial information on the cellular and molecular mechanisms underlying synaptic plasticity in the hippocampus, and this review discusses these mechanisms in brief. Addiction is a chronic relapsing disorder with loss of control over drug taking and drug seeking that is caused by long-lasting memories of drug experience. Relapse to drug use is caused by exposure to context and cues associated with the drug experience, and is a major clinical problem that contributes to the persistence of addiction. This review also briefly discusses some evidence that drugs of abuse alter plasticity in the hippocampus, and that development of novel treatment strategies that reverse or prevent drug-induced synaptic alterations in the hippocampus may reduce relapse behaviors associated with addiction.
Collapse
Affiliation(s)
| | - Chitra D. Mandyam
- VA San Diego Healthcare System, San Diego, CA 92161, USA;
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| |
Collapse
|
18
|
Breijyeh Z, Jubeh B, Bufo SA, Karaman R, Scrano L. Cannabis: A Toxin-Producing Plant with Potential Therapeutic Uses. Toxins (Basel) 2021; 13:117. [PMID: 33562446 PMCID: PMC7915118 DOI: 10.3390/toxins13020117] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023] Open
Abstract
For thousands of years, Cannabis sativa has been utilized as a medicine and for recreational and spiritual purposes. Phytocannabinoids are a family of compounds that are found in the cannabis plant, which is known for its psychotogenic and euphoric effects; the main psychotropic constituent of cannabis is Δ9-tetrahydrocannabinol (Δ9-THC). The pharmacological effects of cannabinoids are a result of interactions between those compounds and cannabinoid receptors, CB1 and CB2, located in many parts of the human body. Cannabis is used as a therapeutic agent for treating pain and emesis. Some cannabinoids are clinically applied for treating chronic pain, particularly cancer and multiple sclerosis-associated pain, for appetite stimulation and anti-emesis in HIV/AIDS and cancer patients, and for spasticity treatment in multiple sclerosis and epilepsy patients. Medical cannabis varies from recreational cannabis in the chemical content of THC and cannabidiol (CBD), modes of administration, and safety. Despite the therapeutic effects of cannabis, exposure to high concentrations of THC, the main compound that is responsible for most of the intoxicating effects experienced by users, could lead to psychological events and adverse effects that affect almost all body systems, such as neurological (dizziness, drowsiness, seizures, coma, and others), ophthalmological (mydriasis and conjunctival hyperemia), cardiovascular (tachycardia and arterial hypertension), and gastrointestinal (nausea, vomiting, and thirst), mainly associated with recreational use. Cannabis toxicity in children is more concerning and can cause serious adverse effects such as acute neurological symptoms (stupor), lethargy, seizures, and even coma. More countries are legalizing the commercial production and sale of cannabis for medicinal use, and some for recreational use as well. Liberalization of cannabis laws has led to increased incidence of toxicity, hyperemesis syndrome, lung disease cardiovascular disease, reduced fertility, tolerance, and dependence with chronic prolonged use. This review focuses on the potential therapeutic effects of cannabis and cannabinoids, as well as the acute and chronic toxic effects of cannabis use on various body systems.
Collapse
Affiliation(s)
- Zeinab Breijyeh
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem Abu Dis P144, Palestine; (Z.B.); (B.J.)
| | - Buthaina Jubeh
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem Abu Dis P144, Palestine; (Z.B.); (B.J.)
| | - Sabino A. Bufo
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
- Department of Geography, Environmental Management & Energy Studies, University of Johannesburg, Johannesburg 2092, South Africa
| | - Rafik Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem Abu Dis P144, Palestine; (Z.B.); (B.J.)
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
| | - Laura Scrano
- Department of European Cultures (DICEM), University of Basilicata, 75100 Matera, Italy;
| |
Collapse
|
19
|
Carvalhais LG, Martinho VC, Ferreiro E, Pinheiro PS. Unraveling the Nanoscopic Organization and Function of Central Mammalian Presynapses With Super-Resolution Microscopy. Front Neurosci 2021; 14:578409. [PMID: 33584169 PMCID: PMC7874199 DOI: 10.3389/fnins.2020.578409] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/03/2020] [Indexed: 12/22/2022] Open
Abstract
The complex, nanoscopic scale of neuronal function, taking place at dendritic spines, axon terminals, and other minuscule structures, cannot be adequately resolved using standard, diffraction-limited imaging techniques. The last couple of decades saw a rapid evolution of imaging methods that overcome the diffraction limit imposed by Abbe's principle. These techniques, including structured illumination microscopy (SIM), stimulated emission depletion (STED), photo-activated localization microscopy (PALM), and stochastic optical reconstruction microscopy (STORM), among others, have revolutionized our understanding of synapse biology. By exploiting the stochastic nature of fluorophore light/dark states or non-linearities in the interaction of fluorophores with light, by using modified illumination strategies that limit the excitation area, these methods can achieve spatial resolutions down to just a few tens of nm or less. Here, we review how these advanced imaging techniques have contributed to unprecedented insight into the nanoscopic organization and function of mammalian neuronal presynapses, revealing new organizational principles or lending support to existing views, while raising many important new questions. We further discuss recent technical refinements and newly developed tools that will continue to expand our ability to delve deeper into how synaptic function is orchestrated at the nanoscopic level.
Collapse
Affiliation(s)
- Lia G Carvalhais
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Vera C Martinho
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Elisabete Ferreiro
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Paulo S Pinheiro
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
20
|
Neuromolecular Mechanisms of Cannabis Action. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1264:15-28. [PMID: 33332001 DOI: 10.1007/978-3-030-57369-0_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Most of our current understanding of the neuromolecular mechanisms of Cannabis action focusses on two plant cannabinoids, THC and CBD. THC acts primarily through presynaptic CB cannabinoid receptors to regulate neurotransmitter release in the brain, spinal cord and peripheral nerves. CBD action, on the other hand, is probably mediated through multiple molecular targets.
Collapse
|
21
|
Soriano D, Vacotto M, Brusco A, Caltana L. Neuronal and synaptic morphological alterations in the hippocampus of cannabinoid receptor type 1 knockout mice. J Neurosci Res 2020; 98:2245-2262. [DOI: 10.1002/jnr.24694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/28/2020] [Accepted: 06/27/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Delia Soriano
- Universidad de Buenos Aires. Facultad de Medicina. 1° Unidad Académica del Departamento de Histología, Embriología, Biología Celular y Genética. Buenos Aires. Argentina. Buenos Aires Argentina
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis (IBCN) CONICET‐Universidad de Buenos Aires Buenos Aires Argentina
| | - Marina Vacotto
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis (IBCN) CONICET‐Universidad de Buenos Aires Buenos Aires Argentina
| | - Alicia Brusco
- Universidad de Buenos Aires. Facultad de Medicina. 1° Unidad Académica del Departamento de Histología, Embriología, Biología Celular y Genética. Buenos Aires. Argentina. Buenos Aires Argentina
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis (IBCN) CONICET‐Universidad de Buenos Aires Buenos Aires Argentina
| | - Laura Caltana
- Universidad de Buenos Aires. Facultad de Medicina. 1° Unidad Académica del Departamento de Histología, Embriología, Biología Celular y Genética. Buenos Aires. Argentina. Buenos Aires Argentina
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis (IBCN) CONICET‐Universidad de Buenos Aires Buenos Aires Argentina
| |
Collapse
|
22
|
Kesner AJ, Lovinger DM. Cannabinoids, Endocannabinoids and Sleep. Front Mol Neurosci 2020; 13:125. [PMID: 32774241 PMCID: PMC7388834 DOI: 10.3389/fnmol.2020.00125] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/22/2020] [Indexed: 12/21/2022] Open
Abstract
Sleep is a vital function of the nervous system that contributes to brain and bodily homeostasis, energy levels, cognitive ability, and other key functions of a variety of organisms. Dysfunctional sleep induces neural problems and is a key part of almost all human psychiatric disorders including substance abuse disorders. The hypnotic effects of cannabis have long been known and there is increasing use of phytocannabinoids and other formulations as sleep aids. Thus, it is crucial to gain a better understanding of the neurobiological basis of cannabis drug effects on sleep, as well as the role of the endogenous cannabinoid system in sleep physiology. In this review article, we summarize the current state of knowledge concerning sleep-related endogenous cannabinoid function derived from research on humans and rodent models. We also review information on acute and chronic cannabinoid drug effects on sleep in these organisms, and molecular mechanisms that may contribute to these effects. We point out the potential benefits of acute cannabinoids for sleep improvement, but also the potential sleep-disruptive effects of withdrawal following chronic cannabinoid drug use. Prescriptions for future research in this burgeoning field are also provided.
Collapse
Affiliation(s)
- Andrew J Kesner
- Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institute of Health (NIH), Bethesda, MD, United States
- Center on Compulsive Behaviors, Intramural Research Program, National Institute of Health (NIH), Bethesda, MD, United States
| | - David M Lovinger
- Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institute of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
23
|
Prini P, Zamberletti E, Manenti C, Gabaglio M, Parolaro D, Rubino T. Neurobiological mechanisms underlying cannabis-induced memory impairment. Eur Neuropsychopharmacol 2020; 36:181-190. [PMID: 32139186 DOI: 10.1016/j.euroneuro.2020.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/27/2020] [Accepted: 02/17/2020] [Indexed: 01/23/2023]
Abstract
A growing body of literature suggests that cannabis intake can induce memory loss in humans and animals. Besides the recreational use, daily cannabis users may also belong to the ever-increasing population of patients who are administered cannabis as a medicine. As such, they also can experience impairments in memory as a negative side effect of their therapy. Comprehension of the neurobiological mechanisms responsible for such detrimental effects would be therefore of paramount relevance to public health. The investigation of neurobiological mechanisms in humans, despite the progress in the development of imaging technologies that allow the study of brain structure and function, still suffers substantial limitations. Animal models, instead, enable us to establish a causal relationship and thus to better elucidate the neurobiological mechanisms underlying the process under study. In this review, we will attempt to collect the insight coming from animal models about cannabis effects on memory, trying to depict a picture of the neurobiological mechanisms contributing to the development of cognitive deficits following cannabis use.
Collapse
Affiliation(s)
- Pamela Prini
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio VA, Italy
| | - Erica Zamberletti
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio VA, Italy
| | - Cristina Manenti
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio VA, Italy
| | - Marina Gabaglio
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio VA, Italy
| | - Daniela Parolaro
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio VA, Italy; Zardi-Gori Foundation, Milan, Italy
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio VA, Italy.
| |
Collapse
|
24
|
Hwang EK, Lupica CR. Altered Corticolimbic Control of the Nucleus Accumbens by Long-term Δ 9-Tetrahydrocannabinol Exposure. Biol Psychiatry 2020; 87:619-631. [PMID: 31543247 PMCID: PMC7002212 DOI: 10.1016/j.biopsych.2019.07.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/14/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND The decriminalization and legalization of cannabis and the expansion of availability of medical cannabis in North America have led to an increase in cannabis use and the availability of high-potency strains. Cannabis potency is determined by the concentration of Δ9-tetrahydrocannabinol (Δ9-THC), a psychoactive constituent that activates cannabinoid CB1 and CB2 receptors. The use of high-potency cannabis is associated with cannabis use disorder and increased susceptibility to psychiatric illness. The nucleus accumbens (NAc) is part of a brain reward circuit affected by Δ9-THC through modulation of glutamate afferents arising from corticolimbic brain areas implicated in drug addiction and psychiatric disorders. Moreover, brain imaging studies show alterations in corticolimbic and NAc properties in human cannabis users. METHODS Using in vitro electrophysiology and optogenetics, we examined how Δ9-THC alters corticolimbic input to the NAc in rats. RESULTS We found that long-term exposure to Δ9-THC weakens prefrontal cortex glutamate input to the NAc shell and strengthens input from basolateral amygdala and ventral hippocampus. Further, whereas long-term exposure to Δ9-THC had no effect on net strength of glutamatergic input to NAc shell arising from midbrain dopamine neurons, it alters fundamental properties of these synapses. CONCLUSIONS Long-term exposure to Δ9-THC shifts control of the NAc shell from cortical to limbic input, likely contributing to cognitive and psychiatric dysfunction that is associated with cannabis use.
Collapse
|
25
|
Abstract
Given the aging Baby Boomer generation, changes in cannabis legislation, and the growing acknowledgment of cannabis for its therapeutic potential, it is predicted that cannabis use in the older population will escalate. It is, therefore, important to determine the interaction between the effects of cannabis and aging. The aim of this report is to describe the link between cannabis use and the aging brain. Our review of the literature found few and inconsistent empirical studies that directly address the impact of cannabis use on the aging brain. However, research focused on long-term cannabis use points toward cumulative effects on multimodal systems in the brain that are similarly affected during aging. Specifically, the effects of cannabis and aging converge on overlapping networks in the endocannabinoid, opioid, and dopamine systems that may affect functional decline particularly in the hippocampus and prefrontal cortex, which are critical areas for memory and executive functioning. To conclude, despite the limited current knowledge on the potential interactive effects between cannabis and aging, evidence from the literature suggests that cannabis and aging effects are concurrently present across several neurotransmitter systems. There is a great need for future research to directly test the interactions between cannabis and aging.
Collapse
Affiliation(s)
- Hye Bin Yoo
- Center for BrainHealth, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas, USA
| | - Jennifer DiMuzio
- Center for BrainHealth, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas, USA
| | - Francesca M Filbey
- Center for BrainHealth, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas, USA
| |
Collapse
|
26
|
Vozella V, Zibardi C, Ahmed F, Piomelli D. Fast and Sensitive Quantification of Δ 9-Tetrahydrocannabinol and Its Main Oxidative Metabolites by Liquid Chromatography/Tandem Mass Spectrometry. Cannabis Cannabinoid Res 2019; 4:110-123. [PMID: 31236476 PMCID: PMC6590723 DOI: 10.1089/can.2018.0075] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: Few animal studies have evaluated the pharmacological effects of Δ9-tetrahydrocannabinol (THC) in relation to its pharmacokinetic properties. Understanding this relationship is essential, however, if comparisons are to be drawn across conditions-such as sex, age, and route of administration-which are associated with variations in the absorption, metabolism, and distribution of THC. As a first step toward addressing this gap, in this report, we describe a rapid, sensitive, and accurate method for the quantification of THC and its main oxidative metabolites, and apply it to representative rodent tissues. Materials and Methods: The sample workup procedure consisted of two steps: bulk protein precipitation with cold acetonitrile (ACN) followed by phospholipid removal by elution through Captiva-Enhanced Matrix Removal cartridges. The liquid chromatography/tandem mass spectrometry (LC/MS-MS) protocol utilized a commercially available C18 reversed-phase column and a simple methanol/water gradient system. The new method was validated following Food and Drug Administration (FDA) guidelines, and was applied to the quantification of THC and its main oxidative metabolites-11-hydroxy-Δ9-tetrahydrocannabinol (11-OH-THC) and 11-nor-9-carboxy-Δ9-tetrahydrocannabinol (11-COOH-THC)-in plasma and brain of mice treated with a single intraperitoneal dose of THC (10 mg/kg). Results: ACN precipitation and column elution effectively depleted matrix constituents-most notably choline-containing phospholipids-which are known to interfere with THC analysis, with average recovery values of >85% for plasma and >80% for brain. The LC conditions yielded baseline separation of all analytes in a total run time of 7 min (including re-equilibration). The 10-point calibration curves showed excellent linearity (R 2>0.99) over a wide range of concentrations (1-1000 pmol/100 μL). Lowest limit of quantification was 2 pmol/100 μL for all analytes, and lowest limits of detection were 0.5 pmol/100 μL for THC and 11-OH-THC, and 1 pmol/100 μL for 11-COOH-THC. Intraday and interday accuracy and precision values were within the FDA-recommended range (±15% of nominal concentration). An application of the method to adult male mice is presented. Conclusions: We present a fast and sensitive method for the analysis of THC, which should facilitate studies aimed at linking the pharmacokinetics and pharmacodynamics of this compound in animal models.
Collapse
Affiliation(s)
- Valentina Vozella
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California
| | - Cristina Zibardi
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California
| | - Faizy Ahmed
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California
- Department of Biological Chemistry, University of California, Irvine, Irvine, California
- Department of Pharmacology, University of California, Irvine, Irvine, California
- Center for the Study of Cannabis, University of California, Irvine, Irvine, California
| |
Collapse
|
27
|
Capsaicin inhibits the function of α 7-nicotinic acetylcholine receptors expressed in Xenopus oocytes and rat hippocampal neurons. Eur J Pharmacol 2019; 857:172411. [PMID: 31152699 DOI: 10.1016/j.ejphar.2019.172411] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 11/21/2022]
Abstract
Capsaicin is a naturally occurring alkaloid derived from Chili peppers fruits. Using the two-electrode voltage-clamp technique in Xenopus oocyte expression system, actions of capsaicin on the functional properties of α7 subunit of the human nicotinic acetylcholine (α7 nACh) receptor were investigated. Ion currents activated by ACh (100 μM) were reversibly inhibited with an IC50 value of 8.6 μM. Inhibitory actions of capsaicin was independent of membrane potential. Furthermore, Ca2+-dependent Cl- channels expressed endogenously in oocytes were not involved in inhibitory actions of capsaicin. In addition, increasing the ACh concentrations could not reverse the inhibitory effects of capsaicin. Importantly, specific binding of [125I] α-bungarotoxin remained unaltered by capsaicin suggesting that its effect is noncompetitive. Whole cell patch-clamp technique was performed in CA1 stratum radiatum interneurons of rat hippocampal slices. Ion currents induced by choline, a selective-agonist of α7-receptor, were reversibly inhibited by 10 min bath application of capsaicin (10 μM). Collectively, results of our investigation indicate that the function of the α7-nACh receptor expressed in Xenopus oocytes and in hippocampal interneurons are inhibited by capsaicin.
Collapse
|
28
|
Pinky PD, Bloemer J, Smith WD, Moore T, Hong H, Suppiramaniam V, Reed MN. Prenatal cannabinoid exposure and altered neurotransmission. Neuropharmacology 2019; 149:181-194. [PMID: 30771373 DOI: 10.1016/j.neuropharm.2019.02.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/18/2019] [Accepted: 02/12/2019] [Indexed: 11/26/2022]
Abstract
Marijuana is one of the most commonly used illicit drugs worldwide. In addition, use of synthetic cannabinoids is increasing, especially among adolescents and young adults. Although human studies have shown that the use of marijuana during pregnancy leads to adverse behavioral effects, such as deficiencies in attention and executive function in affected offspring, the rate of marijuana use among pregnant women is steadily increasing. Various aspects of human behavior including emotion, learning, and memory are dependent on complex interactions between multiple neurotransmitter systems that are especially vulnerable to alterations during the developmental period. Thus, exploration of neurotransmitter changes in response to prenatal cannabinoid exposure is crucial to develop an understanding of how homeostatic imbalance and various long-term neurobehavioral deficits manifest following the abuse of marijuana or other synthetic cannabinoids during pregnancy. Current literature confirms that vast alterations to neurotransmitter systems are present following prenatal cannabinoid exposure, and many of these alterations within the brain are region specific, time-dependent, and sexually dimorphic. In this review, we aim to provide a summary of observed changes to various neurotransmitter systems following cannabinoid exposure during pregnancy and to draw possible correlations to reported behavioral alterations in affected offspring.
Collapse
Affiliation(s)
- Priyanka D Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Warren D Smith
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Timothy Moore
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| | - Hao Hong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA.
| | - Miranda N Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA.
| |
Collapse
|
29
|
Borsoi M, Manduca A, Bara A, Lassalle O, Pelissier-Alicot AL, Manzoni OJ. Sex Differences in the Behavioral and Synaptic Consequences of a Single in vivo Exposure to the Synthetic Cannabimimetic WIN55,212-2 at Puberty and Adulthood. Front Behav Neurosci 2019; 13:23. [PMID: 30890922 PMCID: PMC6411818 DOI: 10.3389/fnbeh.2019.00023] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/30/2019] [Indexed: 01/08/2023] Open
Abstract
Heavy cannabis consumption among adolescents is associated with significant and lasting neurobiological, psychological and health consequences that depend on the age of first use. Chronic exposure to cannabinoid agonists during the perinatal period or adolescence alters social behavior and prefrontal cortex (PFC) activity in adult rats. However, sex differences on social behavior as well as PFC synaptic plasticity after acute cannabinoid activation remain poorly explored. Here, we determined that the consequences of a single in vivo exposure to the synthetic cannabimimetic WIN55,212-2 differently affected PFC neuronal and synaptic functions after 24 h in male and female rats during the pubertal and adulthood periods. During puberty, single cannabinoid exposure (SCE) reduced play behavior in females but not males. In contrast, the same treatment impaired sociability in both sexes at adulthood. General exploration and memory recognition remained normal at both ages and both sexes. At the synaptic level, SCE ablated endocannabinoid-mediated synaptic plasticity in the PFC of females of both ages and heightened excitability of PFC pyramidal neurons at adulthood, while males were spared. In contrast, cannabinoid exposure was associated with impaired long-term potentiation (LTP) specifically in adult males. Together, these data indicate behavioral and synaptic sex differences in response to a single in vivo exposure to cannabinoid at puberty and adulthood.
Collapse
Affiliation(s)
- Milene Borsoi
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France.,Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Indiana University, Marseille, France
| | - Antonia Manduca
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France.,Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Indiana University, Marseille, France
| | - Anissa Bara
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France.,Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Indiana University, Marseille, France
| | - Olivier Lassalle
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France.,Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Indiana University, Marseille, France
| | - Anne-Laure Pelissier-Alicot
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France.,Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Indiana University, Marseille, France.,Assistance Publique Hôpitaux de Marseille (APHM), CHU Conception, Service de Psychiatrie, Marseille, France.,Assistance Publique Hôpitaux de Marseille (APHM), CHU Timone Adultes, Service de Médecine Légale, Marseille, France
| | - Olivier J Manzoni
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France.,Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Indiana University, Marseille, France
| |
Collapse
|
30
|
Endocannabinoid System and Alcohol Abuse Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1162:89-127. [PMID: 31332736 DOI: 10.1007/978-3-030-21737-2_6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Δ9-tetrahydrocannabinol (Δ9-THC), the primary active component in Cannabis sativa preparations such as hashish and marijuana, signals by binding to cell surface receptors. Two types of receptors have been cloned and characterized as cannabinoid (CB) receptors. CB1 receptors (CB1R) are ubiquitously present in the central nervous system (CNS) and are present in both inhibitory interneurons and excitatory neurons at the presynaptic terminal. CB2 receptors (CB2R) are demonstrated in microglial cells, astrocytes, and several neuron subpopulations and are present in both pre- and postsynaptic terminals. The majority of studies on these receptors have been conducted in the past two and half decades after the identification of the molecular constituents of the endocannabinoid (eCB) system that started with the characterization of CB1R. Subsequently, the seminal discovery was made, which suggested that alcohol (ethanol) alters the eCB system, thus establishing the contribution of the eCB system in the motivation to consume ethanol. Several preclinical studies have provided evidence that CB1R significantly contributes to the motivational and reinforcing properties of ethanol and that the chronic consumption of ethanol alters eCB transmitters and CB1R expression in the brain nuclei associated with addiction pathways. Additionally, recent seminal studies have further established the role of the eCB system in the development of ethanol-induced developmental disorders, such as fetal alcohol spectrum disorders (FASD). These results are augmented by in vitro and ex vivo studies, showing that acute and chronic treatment with ethanol produces physiologically relevant alterations in the function of the eCB system during development and in the adult stage. This chapter provides a current and comprehensive review of the literature concerning the role of the eCB system in alcohol abuse disorders (AUD).
Collapse
|
31
|
Secci ME, Mascia P, Sagheddu C, Beggiato S, Melis M, Borelli AC, Tomasini MC, Panlilio LV, Schindler CW, Tanda G, Ferré S, Bradberry CW, Ferraro L, Pistis M, Goldberg SR, Schwarcz R, Justinova Z. Astrocytic Mechanisms Involving Kynurenic Acid Control Δ 9-Tetrahydrocannabinol-Induced Increases in Glutamate Release in Brain Reward-Processing Areas. Mol Neurobiol 2018; 56:3563-3575. [PMID: 30151725 DOI: 10.1007/s12035-018-1319-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/14/2018] [Indexed: 12/27/2022]
Abstract
The reinforcing effects of Δ9-tetrahydrocannabinol (THC) in rats and monkeys, and the reinforcement-related dopamine-releasing effects of THC in rats, can be attenuated by increasing endogenous levels of kynurenic acid (KYNA) through systemic administration of the kynurenine 3-monooxygenase inhibitor, Ro 61-8048. KYNA is a negative allosteric modulator of α7 nicotinic acetylcholine receptors (α7nAChRs) and is synthesized and released by astroglia, which express functional α7nAChRs and cannabinoid CB1 receptors (CB1Rs). Here, we tested whether these presumed KYNA autoreceptors (α7nAChRs) and CB1Rs regulate glutamate release. We used in vivo microdialysis and electrophysiology in rats, RNAscope in situ hybridization in brain slices, and primary culture of rat cortical astrocytes. Acute systemic administration of THC increased extracellular levels of glutamate in the nucleus accumbens shell (NAcS), ventral tegmental area (VTA), and medial prefrontal cortex (mPFC). THC also reduced extracellular levels of KYNA in the NAcS. These THC effects were prevented by administration of Ro 61-8048 or the CB1R antagonist, rimonabant. THC increased the firing activity of glutamatergic pyramidal neurons projecting from the mPFC to the NAcS or to the VTA in vivo. These effects were averted by pretreatment with Ro 61-8048. In vitro, THC elicited glutamate release from cortical astrocytes (on which we demonstrated co-localization of the CB1Rs and α7nAChR mRNAs), and this effect was prevented by KYNA and rimonabant. These results suggest a key role of astrocytes in interactions between the endocannabinoid system, kynurenine pathway, and glutamatergic neurotransmission, with ramifications for the pathophysiology and treatment of psychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria E Secci
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Paola Mascia
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Sarah Beggiato
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Andrea C Borelli
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Maria C Tomasini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Leigh V Panlilio
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Charles W Schindler
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Gianluigi Tanda
- Molecular Targets and Medications Discovery Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Sergi Ferré
- Molecular Targets and Medications Discovery Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Charles W Bradberry
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
- National Research Council of Italy (CNR), Section of Cagliari, Neuroscience Institute, Monserrato, Italy
| | - Steven R Goldberg
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zuzana Justinova
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA.
| |
Collapse
|
32
|
Lovell ME, Bruno R, Johnston J, Matthews A, McGregor I, Allsop DJ, Lintzeris N. Cognitive, physical, and mental health outcomes between long-term cannabis and tobacco users. Addict Behav 2018; 79:178-188. [PMID: 29291509 DOI: 10.1016/j.addbeh.2017.12.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/17/2017] [Accepted: 12/07/2017] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Cannabis intoxication adversely affects health, yet persistent effects following short-term abstinence in long-term cannabis users are unclear. This matched-subjects, cross-sectional study compared health outcomes of long-term cannabis and long-term tobacco-only users, relative to population norms. METHODS Nineteen long-term (mean 32.3years of use, mean age 55.7years), abstinent (mean 15h) cannabis users and 16 long-term tobacco users (mean 37.1years of use, mean age 52.9years), matched for age, educational attainment, and lifetime tobacco consumption, were compared on measures of learning and memory, response inhibition, information-processing, sustained attention, executive control, and mental and physical health. RESULTS Cannabis users exhibited poorer overall learning and delayed recall and greater interference and forgetting than tobacco users, and exhibited poorer recall than norms. Inhibition and executive control were similar between groups, but cannabis users had slower reaction times during information processing and sustained attention tasks. Cannabis users had superior health satisfaction and psychological, somatic, and general health than tobacco users and had similar mental and physical health to norms whilst tobacco users had greater stress, role limitations from emotional problems, and poorer health satisfaction. CONCLUSIONS Long-term cannabis users may exhibit deficits in some cognitive domains despite short-term abstinence and may therefore benefit from interventions to improve cognitive performance. Tobacco alone may contribute to adverse mental and physical health outcomes, which requires appropriate control in future studies.
Collapse
Affiliation(s)
- M E Lovell
- School of Medicine (Psychology), University of Tasmania, Hobart, Tasmania 7000, Australia.
| | - R Bruno
- School of Medicine (Psychology), University of Tasmania, Hobart, Tasmania 7000, Australia
| | - J Johnston
- University Centre for Rural Health - North Coast, University of Sydney, Lismore, New South Wales 2480, Australia
| | - A Matthews
- School of Medicine (Psychology), University of Tasmania, Hobart, Tasmania 7000, Australia
| | - I McGregor
- Lambert Initiative for Cannabinoid Therapeutics, School of Psychology, Brain and Mind Centre, University of Sydney, New South Wales 2006, Australia
| | - D J Allsop
- Lambert Initiative for Cannabinoid Therapeutics, School of Psychology, Brain and Mind Centre, University of Sydney, New South Wales 2006, Australia
| | - N Lintzeris
- Discipline of Addictive Medicine, Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia; The Langton Centre, South East Sydney Local Health District (SESLHD), Drug and Alcohol Services, 2010, Australia
| |
Collapse
|
33
|
Myers AM, Siegele PB, Foss JD, Tuma RF, Ward SJ. Single and combined effects of plant-derived and synthetic cannabinoids on cognition and cannabinoid-associated withdrawal signs in mice. Br J Pharmacol 2018; 176:1552-1567. [PMID: 29338068 DOI: 10.1111/bph.14147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/04/2017] [Accepted: 10/11/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND PURPOSE It has been suggested that the non-euphorogenic phytocannabinoid cannabidiol (CBD) can ameliorate adverse effects of Δ9 -tetrahydrocannabinol (THC). We determined whether CBD ameliorates cognitive deficits and withdrawal signs induced by cannabinoid CB1 /CB2 receptor agonists or produces these pharmacological effects on its own. EXPERIMENTAL APPROACH The effects of THC or the CB1 /CB2 receptor full agonist WIN55212 alone, CBD alone or their combination were tested across a range of doses. Cognitive effects were assessed in C57BL/6 mice in a conditional discrimination task and in the Barnes maze. Cannabinoid withdrawal signs were assessed following precipitated withdrawal by acute administration of the CB1 receptor antagonist SR141716, the 5-HT1A receptor antagonist WAY100635, the TRPV1 receptor antagonist capsazepine or the adenosine A2A receptor antagonist SCH58261. KEY RESULTS THC produced significant motor and cognitive impairment in the Barnes maze task, none of which were attenuated by the addition of CBD. CBD alone did not affect cognitive performance. Precipitation of withdrawal signs by SR141716 occurred in mice chronically treated with THC or WIN55,212. These withdrawal signs were not attenuated by addition of chronic CBD. Chronic treatment with CBD alone did not induce withdrawal signs precipitated by SR141716 or WAY100635. Chronic CBD treatment also produced anxiolysis, which was not altered by attempting to precipitate withdrawal-induced anxiety with a range of antagonists. CONCLUSIONS AND IMPLICATIONS CBD as a monotherapy may prove to be a safer pharmacological agent, than CB1 receptor agonists alone or in combination with CBD, for the treatment of several disorders. LINKED ARTICLES This article is part of a themed section on 8th European Workshop on Cannabinoid Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.10/issuetoc.
Collapse
Affiliation(s)
- Alyssa M Myers
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Patrick B Siegele
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jeffrey D Foss
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Ronald F Tuma
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Sara Jane Ward
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
34
|
Profound deficits in hippocampal synaptic plasticity after traumatic brain injury and seizure is ameliorated by prophylactic levetiracetam. Oncotarget 2018; 9:11515-11527. [PMID: 29545916 PMCID: PMC5837755 DOI: 10.18632/oncotarget.23923] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/29/2017] [Indexed: 11/25/2022] Open
Abstract
Aim To determine the precise effects of post-traumatic seizure activity on hippocampal processes, we induced seizures at various intervals after traumatic brain injury (TBI) and analyzed plasticity at CA1 Schaffer collateral synapses. Material and Methods Rats were initially separated into two groups; one exposed solely to fluid percussion injury (FPI) at 2 Psi and the other only receiving kainic acid (KA)-induced seizures without FPI. Electrophysiological (ePhys) studies including paired-pulse stimulation for short-term presynaptic plasticity and long-term potentiation (LTP) of CA1 Schaffer collateral synapses of the hippocampus for post-synaptic function survey were followed at post-event 1 hour, 3 and 7 days respectively. Additional rats were exposed to three seizures at weekly intervals starting 1 week or 2 weeks after TBI and compared with seizures without TBI, TBI without seizures, and uninjured animals. An additional group placed under the same control variables were treated with levetiracetam prior to seizure induction. The ePhys studies related to post-TBI induced seizures were also followed in these additional groups. Results Seizures affected the short- and long-term synaptic plasticity of the hippocampal CA3-CA1 pathway. FPI itself suppressed LTP and field excitatory post synaptic potentials (fEPSP) in the CA1 Schaffer collateral synapses; KA-induced seizures that followed FPI further suppressed synaptic plasticity. The impairments in both short-term presynaptic and long-term plasticity were worse in the rats in which early post-TBI seizures were induced than those in which later post-TBI seizures were induced. Finally, prophylactic infusion of levetiracetam for one week after FPI reduced the synaptic plasticity deficits in early post-TBI seizure animals. Conclusion Our data indicates that synaptic plasticity (i.e., both presynaptic and postsynaptic) suppression occurs in TBI followed by a seizure and that the interval between the TBI and seizure is an important factor in the severity of the resulting deficits. Furthermore, the infusion of prophylactic levetiracetam could partially reverse the suppression of synaptic plasticity.
Collapse
|
35
|
Thujone inhibits the function of α 7-nicotinic acetylcholine receptors and impairs nicotine-induced memory enhancement in one-trial passive avoidance paradigm. Toxicology 2017; 384:23-32. [PMID: 28395994 DOI: 10.1016/j.tox.2017.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 11/21/2022]
Abstract
Effects of thujone, a major ingredient of absinthe, wormwood oil and some herbal medicines, were tested on the function of α7 subunit of the human nicotinic acetylcholine (α7 nACh) receptor expressed in Xenopus oocytes using the two-electrode voltage-clamp technique. Thujone reversibly inhibited ACh (100μM)-induced currents with an IC50 value of 24.7μM. The effect of thujone was not dependent on the membrane potential and did not involve Ca2+-dependent Cl- channels expressed endogenously in oocytes. Inhibition by thujone was not reversed by increasing ACh concentrations. Moreover, specific binding of [125I] α-bungarotoxin was not altered by thujone. Further experiments in SH-EP1 cells expressing human α7 nACh receptor indicated that thujone suppressed choline induced Ca2+ transients in a concentration-dependent manner. In rat hippocampal CA3-dentate gyrus synapses, nicotine-induced enhancement of long-term potentiation was also inhibited by thujone. Furthermore, the results observed in in-vivo one-trial passive avoidance paradigm show that thujone (1.25mg/kg, i.p.) significantly impaired nicotine-induced enhancement of learning and memory in Wistar rats. Collectively, our results indicate that thujone inhibits the function of the α7-nACh receptor and impairs cellular and behavioral correlates of cholinergic modulation of learning and memory.
Collapse
|
36
|
Shiri M, Komaki A, Oryan S, Taheri M, Komaki H, Etaee F. Effects of cannabinoid and vanilloid receptor agonists and their interaction on learning and memory in rats. Can J Physiol Pharmacol 2017; 95:382-387. [DOI: 10.1139/cjpp-2016-0274] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Despite previous findings on the effects of cannabinoid and vanilloid systems on learning and memory, the effects of the combined stimulation of these 2 systems on learning and memory have not been studied. Therefore, in this study, we tested the interactive effects of cannabinoid and vanilloid systems on learning and memory in rats by using passive avoidance learning (PAL) tests. Forty male Wistar rats were divided into the following 4 groups: (1) control (DMSO+saline), (2) WIN55,212–2, (3) capsaicin, and (4) WIN55,212–2 + capsaicin. On test day, capsaicin, a vanilloid receptor type 1 (TRPV1) agonist, or WIN55,212–2, a cannabinoid receptor (CB1/CB2) agonist, or both substances were injected intraperitoneally. Compared to the control group, the group treated with capsaicin (TRPV1 agonist) had better scores in the PAL acquisition and retention test, whereas treatment with WIN55,212–2 (CB1/CB2 agonist) decreased the test scores. Capsaicin partly reduced the effects of WIN55,212–2 on PAL and memory. We conclude that the acute administration of a TRPV1 agonist improves the rats’ cognitive performance in PAL tasks and that a vanilloid-related mechanism may underlie the agonistic effect of WIN55,212–2 on learning and memory.
Collapse
Affiliation(s)
- Mariam Shiri
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahrbanoo Oryan
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoumeh Taheri
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamidreza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Farshid Etaee
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
37
|
Hoffman AF, Lycas MD, Kaczmarzyk JR, Spivak CE, Baumann MH, Lupica CR. Disruption of hippocampal synaptic transmission and long-term potentiation by psychoactive synthetic cannabinoid 'Spice' compounds: comparison with Δ 9 -tetrahydrocannabinol. Addict Biol 2017; 22:390-399. [PMID: 26732435 PMCID: PMC4935655 DOI: 10.1111/adb.12334] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/16/2015] [Accepted: 10/21/2015] [Indexed: 12/16/2022]
Abstract
There has been a marked increase in the availability of synthetic drugs designed to mimic the effects of marijuana. These cannabimimetic drugs, sold illicitly as 'Spice' and related products, are associated with serious medical complications in some users. In vitro studies suggest that synthetic cannabinoids in these preparations are potent agonists at central cannabinoid CB1 receptors (CB1Rs), but few investigations have delineated their cellular effects, particularly in comparison with the psychoactive component of marijuana, Δ9 -tetrahydrocannabinol (Δ9 -THC). We compared the ability of three widely abused synthetic cannabinoids and Δ9 -THC to alter glutamate release and long-term potentiation in the mouse hippocampus. JWH-018 was the most potent inhibitor of hippocampal synaptic transmission (EC50 ~15 nM), whereas its fluoropentyl derivative, AM2201, inhibited synaptic transmission with slightly lower potency (EC50 ~60 nM). The newer synthetic cannabinoid, XLR-11, displayed much lower potency (EC50 ~900 nM) that was similar to Δ9 -THC (EC50 ~700 nM). The effects of all compounds occurred via activation of CB1Rs, as demonstrated by reversal with the selective antagonist/inverse agonist AM251 or the neutral CB1R antagonist PIMSR1. Moreover, AM2201 was without effect in the hippocampus of transgenic mice lacking the CB1R. Hippocampal slices exposed to either synthetic cannabinoids or Δ9 -THC exhibited significantly impaired long-term potentiation (LTP). We find that, compared with Δ9 -THC, the first-generation cannabinoids found in Spice preparations display higher potency, whereas a recent synthetic cannabinoid is roughly equipotent with Δ9 -THC. The disruption of synaptic function by these synthetic cannabinoids is likely to lead to profound impairments in cognitive and behavioral function.
Collapse
Affiliation(s)
- Alexander F. Hoffman
- Electrophysiology Research Section, Cellular Neurobiology BranchNational Institute on Drug Abuse Intramural Research ProgramBaltimoreMDUSA
| | - Matthew D. Lycas
- Electrophysiology Research Section, Cellular Neurobiology BranchNational Institute on Drug Abuse Intramural Research ProgramBaltimoreMDUSA
| | - Jakub R. Kaczmarzyk
- Electrophysiology Research Section, Cellular Neurobiology BranchNational Institute on Drug Abuse Intramural Research ProgramBaltimoreMDUSA
| | - Charles E. Spivak
- Electrophysiology Research Section, Cellular Neurobiology BranchNational Institute on Drug Abuse Intramural Research ProgramBaltimoreMDUSA
| | - Michael H. Baumann
- Designer Drug Research UnitNational Institute on Drug Abuse Intramural Research ProgramBaltimoreMDUSA
| | - Carl R. Lupica
- Electrophysiology Research Section, Cellular Neurobiology BranchNational Institute on Drug Abuse Intramural Research ProgramBaltimoreMDUSA
| |
Collapse
|
38
|
Kutlu MG, Gould TJ. Effects of drugs of abuse on hippocampal plasticity and hippocampus-dependent learning and memory: contributions to development and maintenance of addiction. Learn Mem 2016; 23:515-33. [PMID: 27634143 PMCID: PMC5026208 DOI: 10.1101/lm.042192.116] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/19/2016] [Indexed: 11/25/2022]
Abstract
It has long been hypothesized that conditioning mechanisms play major roles in addiction. Specifically, the associations between rewarding properties of drugs of abuse and the drug context can contribute to future use and facilitate the transition from initial drug use into drug dependency. On the other hand, the self-medication hypothesis of drug abuse suggests that negative consequences of drug withdrawal result in relapse to drug use as an attempt to alleviate the negative symptoms. In this review, we explored these hypotheses and the involvement of the hippocampus in the development and maintenance of addiction to widely abused drugs such as cocaine, amphetamine, nicotine, alcohol, opiates, and cannabis. Studies suggest that initial exposure to stimulants (i.e., cocaine, nicotine, and amphetamine) and alcohol may enhance hippocampal function and, therefore, the formation of augmented drug-context associations that contribute to the development of addiction. In line with the self-medication hypothesis, withdrawal from stimulants, ethanol, and cannabis results in hippocampus-dependent learning and memory deficits, which suggest that an attempt to alleviate these deficits may contribute to relapse to drug use and maintenance of addiction. Interestingly, opiate withdrawal leads to enhancement of hippocampus-dependent learning and memory. Given that a conditioned aversion to drug context develops during opiate withdrawal, the cognitive enhancement in this case may result in the formation of an augmented association between withdrawal-induced aversion and withdrawal context. Therefore, individuals with opiate addiction may return to opiate use to avoid aversive symptoms triggered by the withdrawal context. Overall, the systematic examination of the role of the hippocampus in drug addiction may help to formulate a better understanding of addiction and underlying neural substrates.
Collapse
Affiliation(s)
- Munir Gunes Kutlu
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania 16802, USA
| | - Thomas J Gould
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania 16802, USA
| |
Collapse
|
39
|
The Influence of the CB1 Receptor Ligands on the Schizophrenia-Like Effects in Mice Induced by MK-801. Neurotox Res 2016; 30:658-676. [PMID: 27577742 PMCID: PMC5047950 DOI: 10.1007/s12640-016-9662-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 08/12/2016] [Accepted: 08/16/2016] [Indexed: 11/20/2022]
Abstract
A growing body of psychiatric research has emerged, focusing on the role of endocannabinoid system in psychiatric disorders. For example, the endocannabinoid system, via cannabinoid CB (CB1 and CB2) receptors, is able to control the function of many receptors, such as N-methyl-d-aspartate (NMDA) receptors connected strictly with psychosis or other schizophrenia-associated symptoms. The aim of the present research was to investigate the impact of the CB1 receptor ligands on the symptoms typical for schizophrenia. We provoked psychosis-like effects in mice by an acute administration of NMDA receptor antagonist, MK-801 (0.1–0.6 mg/kg). An acute administration of MK-801 induced psychotic symptoms, manifested in the increase in locomotor activity (hyperactivity), measured in actimeters, as well as the memory impairment, assessed in the passive avoidance task. We revealed that an acute injection of CB1 receptor agonist, oleamide (5–20 mg/kg), had no influence on the short- and long-term memory-related disturbances, as well as on the hyperlocomotion in mice, provoking by an acute MK-801. In turn, an amnestic effects or hyperactivity induced by an acute MK-801 was attenuated by an acute administration of AM 251 (0.25–3 mg/kg), a CB1 receptor antagonist. The present findings confirm that endocannabinoid system is able to modify a variety of schizophrenia-like responses, including the cognitive disturbances and hyperlocomotion in mice. Antipsychotic-like effects induced by CB1 receptor antagonist, obtained in our research, confirm the potential effect of CB1 receptor blockade and could have important therapeutic implications on clinical settings, in the future.
Collapse
|
40
|
Barbieri M, Ossato A, Canazza I, Trapella C, Borelli AC, Beggiato S, Rimondo C, Serpelloni G, Ferraro L, Marti M. Synthetic cannabinoid JWH-018 and its halogenated derivatives JWH-018-Cl and JWH-018-Br impair Novel Object Recognition in mice: Behavioral, electrophysiological and neurochemical evidence. Neuropharmacology 2016; 109:254-269. [PMID: 27346209 DOI: 10.1016/j.neuropharm.2016.06.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/14/2016] [Accepted: 06/22/2016] [Indexed: 01/29/2023]
Abstract
It is well known that an impairment of learning and memory function is one of the major physiological effects caused by natural or synthetic cannabinoid consumption in rodents, nonhuman primates and in humans. JWH-018 and its halogenated derivatives (JWH-018-Cl and JWH-018-Br) are synthetic CB1/CB2 cannabinoid agonists, illegally marketed as "Spice" and "herbal blend" for their Cannabis-like psychoactive effects. In the present study the effects of acute exposure to JWH-018, JWH-018-Cl, JWH-018-Br (JWH-018-R compounds) and Δ(9)-THC (for comparison) on Novel Object Recognition test (NOR) has been investigated in mice. Moreover, to better characterize the effects of JWH-018-R compounds on memory function, in vitro electrophysiological and neurochemical studies in hippocampal preparations have been performed. JWH-018, JWH-018-Cl and JWH-018-Br dose-dependently impaired both short- and long-memory retention in mice (respectively 2 and 24 h after training session). Their effects resulted more potent respect to that evoked by Δ(9)-THC. Moreover, in vitro studies showed as JWH-018-R compounds negatively affected electrically evoked synaptic transmission, LTP and aminoacid (glutamate and GABA) release in hippocampal slices. Behavioral, electrophysiological and neurochemical effects were fully prevented by CB1 receptor antagonist AM251 pretreatment, suggesting a CB1 receptor involvement. These data support the hypothesis that synthetic JWH-018-R compounds, as Δ(9)-THC, impair cognitive function in mice by interfering with hippocampal synaptic transmission and memory mechanisms. This data outline the danger that the use and/or abuse of these synthetic cannabinoids may represent for the cognitive process in human consumer.
Collapse
Affiliation(s)
- M Barbieri
- Department of Medical Sciences, University of Ferrara, Italy
| | - A Ossato
- Department of Life Sciences and Biotechnology (SVeB), University of Ferrara, Italy
| | - I Canazza
- Department of Life Sciences and Biotechnology (SVeB), University of Ferrara, Italy
| | - C Trapella
- Department of Chemistry and Pharmaceutical Sciences, University of Ferrara, Italy
| | - A C Borelli
- Department of Medical Sciences, University of Ferrara, Italy
| | - S Beggiato
- Department of Life Sciences and Biotechnology (SVeB), University of Ferrara, Italy
| | - C Rimondo
- Department of Public Health and Community Medicine, University of Verona, Italy
| | - G Serpelloni
- U.R.I.To.N., Forensic Toxicology Unit, Department of Health Science, University of Florence, Florence, Italy
| | - L Ferraro
- Department of Life Sciences and Biotechnology (SVeB), University of Ferrara, Italy
| | - M Marti
- Department of Life Sciences and Biotechnology (SVeB), University of Ferrara, Italy; Center for Neuroscience and Istituto Nazionale di Neuroscienze, Italy.
| |
Collapse
|
41
|
Taurisano P, Antonucci LA, Fazio L, Rampino A, Romano R, Porcelli A, Masellis R, Colizzi M, Quarto T, Torretta S, Di Giorgio A, Pergola G, Bertolino A, Blasi G. Prefrontal activity during working memory is modulated by the interaction of variation in CB1 and COX2 coding genes and correlates with frequency of cannabis use. Cortex 2016; 81:231-8. [PMID: 27261878 DOI: 10.1016/j.cortex.2016.05.010] [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: 11/11/2015] [Revised: 04/19/2016] [Accepted: 05/13/2016] [Indexed: 02/02/2023]
Abstract
The CB1 cannabinoid receptor is targeted in the brain by endocannabinoids under physiological conditions as well as by delta9-tetrahydrocannabinol under cannabis use. Furthermore, its signaling appears to affect brain cognitive processing. Recent findings highlight a crucial role of cyclooxygenase-2 (COX-2) in the mechanism of intraneuronal CB1 signaling transduction, while others indicate that two single nucleotide polymorphisms (SNPs) (rs1406977 and rs20417) modulate expression of CB1 (CNR1) and COX-2 (PTGS2) coding genes, respectively. Here, our aim was to use fMRI to investigate in healthy humans whether these SNPs interact in modulating prefrontal activity during working memory processing and if this modulation is linked with cannabis use. We recruited 242 healthy subjects genotyped for CNR1 rs1406977 and PTGS2 rs20417 that performed the N-back working memory task during fMRI and were interviewed using the Cannabis Experience Questionnaire (CEQ). We found that the interaction between CNR1 rs1406977 and PTGS2 rs20417 is associated with dorsolateral prefrontal cortex (DLPFC) activity such that specific genotype configurations (CNR1 C carriers/PTGS2 C carriers and CNR1 TT/PTGS2 GG) predict lower cortical response versus others in spite of similar behavioral accuracy. Furthermore, DLPFC activity in the cluster associated with the CNR1 by PTGS2 interaction was negatively correlated with behavioral efficiency and positively correlated with frequency of cannabis use in cannabis users. These results suggest that a genetically modulated balancing of signaling within the CB1-COX-2 pathway may reflect on more or less efficient patterns of prefrontal activity during working memory. Frequency of cannabis use may be a factor for further modulation of CNR1/PTGS2-mediated cortical processing associated with this cognitive process.
Collapse
Affiliation(s)
- Paolo Taurisano
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy; IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy
| | - Linda A Antonucci
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy; Department of Educational Sciences, Psychology and Communication Science, University of Bari 'Aldo Moro', Bari, Italy
| | - Leonardo Fazio
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy; IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy
| | - Antonio Rampino
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - Raffaella Romano
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - Annamaria Porcelli
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - Rita Masellis
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - Marco Colizzi
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Tiziana Quarto
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy; Cognitive Brain Research Unit, Institute of Behavioral Science, University of Helsinki, Finland
| | - Silvia Torretta
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | | | - Giulio Pergola
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - Alessandro Bertolino
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy; pRED, NORD DTA, Hoffman-La Roche, Ltd., Basel, Switzerland.
| | - Giuseppe Blasi
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy.
| |
Collapse
|
42
|
Mismatch Negativity and P50 Sensory Gating in Abstinent Former Cannabis Users. Neural Plast 2016; 2016:6526437. [PMID: 27019754 PMCID: PMC4785272 DOI: 10.1155/2016/6526437] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/20/2016] [Indexed: 01/31/2023] Open
Abstract
Prolonged heavy exposure to cannabis is associated with impaired cognition and brain functional and structural alterations. We recently reported attenuated mismatch negativity (MMN) and altered P50 sensory gating in chronic cannabis users. This study investigated the extent of brain functional recovery (indexed by MMN and P50) in chronic users after cessation of use. Eighteen ex-users (median 13.5 years prior regular use; median 3.5 years abstinence) and 18 nonusers completed (1) a multifeature oddball task with duration, frequency, and intensity deviants and (2) a P50 paired-click paradigm. Trend level smaller duration MMN amplitude and larger P50 ratios (indicative of poorer sensory gating) were observed in ex-users compared to controls. Poorer P50 gating correlated with prior duration of cannabis use. Duration of abstinence was positively correlated with duration MMN amplitude, even after controlling for age and duration of cannabis use. Impaired sensory gating and attenuated MMN amplitude tended to persist in ex-users after prolonged cessation of use, suggesting a lack of full recovery. An association with prolonged duration of prior cannabis use may indicate persistent cannabis-related alterations to P50 sensory gating. Greater reductions in MMN amplitude with increasing abstinence (positive correlation) may be related to either self-medication or an accelerated aging process.
Collapse
|
43
|
Colizzi M, McGuire P, Pertwee RG, Bhattacharyya S. Effect of cannabis on glutamate signalling in the brain: A systematic review of human and animal evidence. Neurosci Biobehav Rev 2016; 64:359-81. [PMID: 26987641 DOI: 10.1016/j.neubiorev.2016.03.010] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/09/2016] [Accepted: 03/11/2016] [Indexed: 01/04/2023]
Abstract
Use of cannabis or delta-9-tetrahydrocannabinol (Δ9-THC), its main psychoactive ingredient, is associated with psychotic symptoms or disorder. However, the neurochemical mechanism that may underlie this psychotomimetic effect is poorly understood. Although dopaminergic dysfunction is generally recognized as the final common pathway in psychosis, evidence of the effects of Δ9-THC or cannabis use on dopaminergic measures in the brain is equivocal. In fact, it is thought that cannabis or Δ9-THC may not act on dopamine firing directly but indirectly by altering glutamate neurotransmission. Here we systematically review all studies examining acute and chronic effects of cannabis or Δ9-THC on glutamate signalling in both animals and man. Limited research carried out in humans tends to support the evidence that chronic cannabis use reduces levels of glutamate-derived metabolites in both cortical and subcortical brain areas. Research in animals tends to consistently suggest that Δ9-THC depresses glutamate synaptic transmission via CB1 receptor activation, affecting glutamate release, inhibiting receptors and transporters function, reducing enzyme activity, and disrupting glutamate synaptic plasticity after prolonged exposure.
Collapse
Affiliation(s)
- Marco Colizzi
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom
| | - Roger G Pertwee
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Sagnik Bhattacharyya
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom.
| |
Collapse
|
44
|
Nazari M, Komaki A, Karamian R, Shahidi S, Sarihi A, Asadbegi M. The interactive role of CB1 and GABAB receptors in hippocampal synaptic plasticity in rats. Brain Res Bull 2016; 120:123-30. [DOI: 10.1016/j.brainresbull.2015.11.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 01/25/2023]
|
45
|
Ruhl T, Moesbauer K, Oellers N, von der Emde G. The endocannabinoid system and associative learning and memory in zebrafish. Behav Brain Res 2015; 290:61-9. [DOI: 10.1016/j.bbr.2015.04.046] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/11/2015] [Accepted: 04/27/2015] [Indexed: 12/19/2022]
|
46
|
Spechler PA, Orr CA, Chaarani B, Kan KJ, Mackey S, Morton A, Snowe MP, Hudson KE, Althoff RR, Higgins ST, Cattrell A, Flor H, Nees F, Banaschewski T, Bokde ALW, Whelan R, Büchel C, Bromberg U, Conrod P, Frouin V, Papadopoulos D, Gallinat J, Heinz A, Walter H, Ittermann B, Gowland P, Paus T, Poustka L, Martinot JL, Artiges E, Smolka MN, Schumann G, Garavan H. Cannabis use in early adolescence: Evidence of amygdala hypersensitivity to signals of threat. Dev Cogn Neurosci 2015; 16:63-70. [PMID: 26347227 PMCID: PMC4801124 DOI: 10.1016/j.dcn.2015.08.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 08/20/2015] [Accepted: 08/22/2015] [Indexed: 12/03/2022] Open
Abstract
Teenagers experimenting with cannabis may be characterized with fMRI. We report a face processing study of cannabis experimenting teenagers. Cannabis experimenting teenagers exhibit greater amygdala reactivity to angry faces. Very low use of cannabis during adolescence may impact healthy emotional development.
Cannabis use in adolescence may be characterized by differences in the neural basis of affective processing. In this study, we used an fMRI affective face processing task to compare a large group (n = 70) of 14-year olds with a history of cannabis use to a group (n = 70) of never-using controls matched on numerous characteristics including IQ, SES, alcohol and cigarette use. The task contained short movies displaying angry and neutral faces. Results indicated that cannabis users had greater reactivity in the bilateral amygdalae to angry faces than neutral faces, an effect that was not observed in their abstinent peers. In contrast, activity levels in the cannabis users in cortical areas including the right temporal-parietal junction and bilateral dorsolateral prefrontal cortex did not discriminate between the two face conditions, but did differ in controls. Results did not change after excluding subjects with any psychiatric symptomology. Given the high density of cannabinoid receptors in the amygdala, our findings suggest cannabis use in early adolescence is associated with hypersensitivity to signals of threat. Hypersensitivity to negative affect in adolescence may place the subject at-risk for mood disorders in adulthood.
Collapse
Affiliation(s)
- Philip A Spechler
- Vermont Center on Behavior and Health, Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05401, United States.
| | - Catherine A Orr
- Vermont Center for Children, Youth, and Families, University of Vermont, Burlington, VT 05401, United States
| | - Bader Chaarani
- Vermont Center on Behavior and Health, Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05401, United States
| | - Kees-Jan Kan
- Vermont Center on Behavior and Health, Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05401, United States
| | - Scott Mackey
- Vermont Center on Behavior and Health, Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05401, United States
| | - Aaron Morton
- Vermont Center on Behavior and Health, Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05401, United States
| | - Mitchell P Snowe
- Vermont Center on Behavior and Health, Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05401, United States
| | - Kelsey E Hudson
- Vermont Center on Behavior and Health, Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05401, United States
| | - Robert R Althoff
- Vermont Center on Behavior and Health, Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05401, United States
| | - Stephen T Higgins
- Vermont Center on Behavior and Health, Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05401, United States
| | - Anna Cattrell
- Institute of Psychiatry, King's College London, United Kingdom; Medical Research Council - Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Kings College London, De Crespigny Park, London, United Kingdom
| | - Herta Flor
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
| | - Frauke Nees
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neurosciences, Trinity College Dublin, Ireland
| | - Tobias Banaschewski
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neurosciences, Trinity College Dublin, Ireland
| | - Robert Whelan
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neurosciences, Trinity College Dublin, Ireland
| | - Christian Büchel
- University Medical Centre Hamburg-Eppendorf, Haus S10, Martinistr. 52, Hamburg, Germany
| | - Uli Bromberg
- Department of Psychiatry, Universite de Montreal, CHU Ste Justine Hospital, Canada
| | - Patricia Conrod
- Institute of Psychiatry, King's College London, United Kingdom; Department of Psychiatry, Universite de Montreal, CHU Ste Justine Hospital, Canada
| | - Vincent Frouin
- Neurospin, Commissariat à l'Energie Atomique, CEA-Saclay Center, Paris, France
| | - Dimitri Papadopoulos
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf (UKE), Martinistrasse 52, 20246 Hamburg, Germany
| | - Jurgen Gallinat
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf (UKE), Martinistrasse 52, 20246 Hamburg, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité, Universitätsmedizin Berlin, Charitéplatz 1, Berlin, Germany
| | - Henrik Walter
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, Berlin, Germany
| | - Bernd Ittermann
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, Berlin, Germany
| | - Penny Gowland
- School of Psychology, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Tomáš Paus
- Rotman Research Institute, Baycrest and Departments of Psychology and Psychiatry, University of Toronto, Toronto, Ontario M6A 2E1, Canada
| | - Luise Poustka
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neurosciences, Trinity College Dublin, Ireland
| | - Jean-Luc Martinot
- Institut National de la Sante et de la Recherche Medicale, INSERM CEAUnit1000, "Imaging & Psychiatry", University Paris Sud, 91400 Orsay, France
| | - Eric Artiges
- Rotman Research Institute, Baycrest and Departments of Psychology and Psychiatry, University of Toronto, Toronto, Ontario M6A 2E1, Canada
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Gunter Schumann
- Institute of Psychiatry, King's College London, United Kingdom; Medical Research Council - Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Kings College London, De Crespigny Park, London, United Kingdom
| | - Hugh Garavan
- Vermont Center on Behavior and Health, Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05401, United States
| | | |
Collapse
|
47
|
Wetherill RR, Jagannathan K, Hager N, Childress AR, Rao H, Franklin TR. Cannabis, Cigarettes, and Their Co-Occurring Use: Disentangling Differences in Gray Matter Volume. Int J Neuropsychopharmacol 2015; 18:pyv061. [PMID: 26045474 PMCID: PMC4648161 DOI: 10.1093/ijnp/pyv061] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 05/25/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Structural magnetic resonance imaging techniques are powerful tools for examining the effects of drug use on the brain. The nicotine and cannabis literature has demonstrated differences between nicotine cigarette smokers and cannabis users compared to controls in brain structure; however, less is known about the effects of co-occurring cannabis and tobacco use. METHODS We used voxel-based morphometry to examine gray matter volume differences between four groups: (1) cannabis-dependent individuals who do not smoke tobacco (Cs); (2) cannabis-dependent individuals who smoke tobacco (CTs); (3) cannabis-naïve, nicotine-dependent individuals who smoke tobacco (Ts); and (4) healthy controls (HCs). We also explored associations between gray matter volume and measures of cannabis and tobacco use. RESULTS A significant group effect was observed in the left putamen, thalamus, right precentral gyrus, and left cerebellum. Compared to HCs, the Cs, CTs, and Ts exhibited larger gray matter volumes in the left putamen. Cs also had larger gray matter volume than HCs in the right precentral gyrus. Cs and CTs exhibited smaller gray matter volume than HCs in the thalamus, and CTs and Ts had smaller left cerebellar gray matter volume than HCs. CONCLUSIONS This study extends previous research that independently examined the effects of cannabis or tobacco use on brain structure by including an examination of co-occurring cannabis and tobacco use, and provides evidence that cannabis and tobacco exposure are associated with alterations in brain regions associated with addiction.
Collapse
Affiliation(s)
- Reagan R Wetherill
- University of Pennsylvania, Department of Psychiatry, Philadelphia, PA (Drs Wetherill, Jagannathan, Childress, Rao, and Franklin, and Mr Hager).
| | - Kanchana Jagannathan
- University of Pennsylvania, Department of Psychiatry, Philadelphia, PA (Drs Wetherill, Jagannathan, Childress, Rao, and Franklin, and Mr Hager)
| | - Nathan Hager
- University of Pennsylvania, Department of Psychiatry, Philadelphia, PA (Drs Wetherill, Jagannathan, Childress, Rao, and Franklin, and Mr Hager)
| | - Anna Rose Childress
- University of Pennsylvania, Department of Psychiatry, Philadelphia, PA (Drs Wetherill, Jagannathan, Childress, Rao, and Franklin, and Mr Hager)
| | - Hengyi Rao
- University of Pennsylvania, Department of Psychiatry, Philadelphia, PA (Drs Wetherill, Jagannathan, Childress, Rao, and Franklin, and Mr Hager)
| | - Teresa R Franklin
- University of Pennsylvania, Department of Psychiatry, Philadelphia, PA (Drs Wetherill, Jagannathan, Childress, Rao, and Franklin, and Mr Hager)
| |
Collapse
|
48
|
Scientific Opinion on the risks for human health related to the presence of tetrahydrocannabinol (THC) in milk and other food of animal origin. EFSA J 2015. [DOI: 10.2903/j.efsa.2015.4141] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
49
|
Stern CAJ, Gazarini L, Vanvossen AC, Zuardi AW, Galve-Roperh I, Guimaraes FS, Takahashi RN, Bertoglio LJ. Δ9-Tetrahydrocannabinol alone and combined with cannabidiol mitigate fear memory through reconsolidation disruption. Eur Neuropsychopharmacol 2015; 25:958-65. [PMID: 25799920 DOI: 10.1016/j.euroneuro.2015.02.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 12/26/2014] [Accepted: 02/06/2015] [Indexed: 12/31/2022]
Abstract
Δ(9)-tetrahydrocannabinol (THC) and cannabidiol (CBD) are the major constituents of the Cannabis sativa plant, which is frequently consumed by subjects exposed to life-threatening situations to relief their symptomatology. It is still unknown, however, whether THC could also affect the maintenance of an aversive memory formed at that time when taken separately and/or in conjunction with CBD. The present study sought to investigate this matter at a preclinical level. We report that THC (0.3-10mg/kg, i.p.) was able to disrupt the reconsolidation of a contextual fear memory, resulting in reduced conditioned freezing expression for over 22 days. This effect was dependent on activation of cannabinoid type-1 receptors located in prelimbic subregion of the medial prefrontal cortex and on memory retrieval/reactivation. Since CBD may counteract the negative psychotropic effects induced by THC and has been shown to be a reconsolidation blocker, we then investigated and demonstrated that associating sub-effective doses of these two compounds was equally effective in attenuating fear memory maintenance in an additive fashion and in a dose ratio of 10 to 1, which contrasts with that commonly found in C. sativa recreational samples. Of note, neither THC alone nor CBD plus THC interfered with anxiety-related behaviors and locomotor activity, as assessed in the elevated plus-maze test, at a time point coinciding with that used to evaluate their effects on memory reconsolidation. Altogether, present findings suggest a potential therapeutic value of using THC and/or CBD to mitigate a dysfunctional aversive memory through reconsolidation disruption in post-traumatic stress disorder patients.
Collapse
Affiliation(s)
- Cristina A J Stern
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Lucas Gazarini
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Ana C Vanvossen
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Antonio W Zuardi
- Department of Neurology, Psychiatry and Medical Psychology, University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Ismael Galve-Roperh
- Department of Biochemistry and Molecular Biology I, Complutense University, Madrid, Spain; Centro de Investigación Biomedica en red en Enfermedades Neurodegenerativas, Madrid, Spain
| | | | - Reinaldo N Takahashi
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Leandro J Bertoglio
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil.
| |
Collapse
|
50
|
Abstract
The endocannabinoid system negatively regulates the release of various neurotransmitters in an activity-dependent manner, thereby influencing the excitability of neuronal circuits. In the hippocampus, cannabinoid type 1 (CB1) receptor is present on both GABAergic and glutamatergic axon terminals. CB1 receptor-deficient mice were previously shown to have increased hippocampal long-term potentiation (LTP). In this study, we have investigated the consequences of cell-type-specific deletion of the CB1 receptor on the induction of hippocampal LTP and on CA1 pyramidal cell morphology. Deletion of CB1 receptor in GABAergic neurons in GABA-CB1-KO mice leads to a significantly decreased hippocampal LTP compared with WT controls. Concomitantly, CA1 pyramidal neurons have a significantly reduced dendritic branching both on the apical and on the basal dendrites. Moreover, the average spine density on the apical dendrites of CA1 pyramidal neurons is significantly diminished. In contrast, in mice lacking CB1 receptor in glutamatergic cells (Glu-CB1-KO), hippocampal LTP is significantly enhanced and CA1 pyramidal neurons show an increased branching and an increased spine density in the apical dendritic region. Together, these results indicate that the CB1 receptor signaling system both on inhibitory and excitatory neurons controls functional and structural synaptic plasticity of pyramidal neurons in the hippocampal CA1 region to maintain an appropriate homeostatic state upon neuronal activation. Consequently, if the CB1 receptor is lost in either neuronal population, an allostatic shift will occur leading to a long-term dysregulation of neuronal functions.
Collapse
|