1
|
Martínez‐Torres A, Morán J. CB1 Receptor Activation Provides Neuroprotection in an Animal Model of Glutamate-Induced Excitotoxicity Through a Reduction of NOX-2 Activity and Oxidative Stress. CNS Neurosci Ther 2024; 30:e70099. [PMID: 39496572 PMCID: PMC11534500 DOI: 10.1111/cns.70099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/27/2024] [Accepted: 10/14/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Excitotoxicity is a process in which NADPH oxidase-2 (NOX-2) plays a pivotal role in the generation of reactive oxygen species (ROS). Oxidative stress influences the expression of Aquaporin 4 (AQP4), a water channel implicated in blood-brain barrier (BBB) permeability and edema formation. The endocannabinoid system is widely distributed in the brain, particularly through the cannabinoid receptor type 1 (CB1) and type 2 (CB2), which have been shown to have a neuroprotective function in brain injury. Given the significant involvement of NOX-2 in ROS production during excitotoxicity, our research aims to assess the participation of NOX-2 in the neuroprotective effect of the cannabinoid receptor agonist WIN55,212-2 against glutamate-induced excitotoxicity damage in the striatum using in vivo model. METHODS Wild-type mice (C57BL/6) and NOX-2 KO (gp91Cybbtm1Din/J) were stereotactically injected in the striatum with monosodium glutamate or vehicle. Subsequently, a group of mice was administered an intraperitoneal dose of WIN55,212-2, AM251, or AM251/WIN55,212-2 following the intracerebral injection. Motor activity was assessed, and the lesion was examined through histological sections stained with cresyl violet. Additionally, brain water content and Evans blue assay were conducted. The activity of NOX was quantified, and the protein expression of CB1, gp91phox, AQP4, Iba-1, TNF-α, and NF-κB was analyzed using Western blot. Furthermore, ROS formation was measured through the DHE assay. RESULTS The activation of the endocannabinoid receptors demonstrated a neuroprotective response during excitotoxicity, meditated by NOX-2. The reduction in ROS production led to a decrease in neuroinflammation, and AQP4 expression, resulting in reduced edema formation, and BBB permeability. CONCLUSIONS During excitotoxic damage, WIN55,212-2 inhibits NOX-2-induced ROS production, reducing brain injury.
Collapse
Affiliation(s)
- Ari Misael Martínez‐Torres
- División de Neurociencias, Instituto de Fisiología CelularUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología CelularUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| |
Collapse
|
2
|
Navarri X, Robertson DN, Charfi I, Wünnemann F, Sâmia Fernandes do Nascimento A, Trottier G, Leclerc S, Andelfinger GU, Di Cristo G, Richer L, Pike GB, Pausova Z, Piñeyro G, Paus T. Cells and Molecules Underpinning Cannabis-Related Variations in Cortical Thickness during Adolescence. J Neurosci 2024; 44:e2256232024. [PMID: 39214708 PMCID: PMC11466068 DOI: 10.1523/jneurosci.2256-23.2024] [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: 12/04/2023] [Revised: 06/05/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
During adolescence, cannabis experimentation is common, and its association with interindividual variations in brain maturation well studied. Cellular and molecular underpinnings of these system-level relationships are, however, unclear. We thus conducted a three-step study. First, we exposed adolescent male mice to Δ-9-tetrahydrocannabinol (THC) or a synthetic cannabinoid WIN 55,212-2 (WIN) and assessed differentially expressed genes (DEGs), spine numbers, and dendritic complexity in their frontal cortex. Second, in human (male) adolescents, we examined group differences in cortical thickness in 34 brain regions, using magnetic resonance imaging, between those who experimented with cannabis before age 16 (n = 140) and those who did not (n = 327). Finally, we correlated spatially these group differences with gene expression of human homologs of mouse-identified DEGs. The spatial expression of 13 THC-related human homologs of DEGs correlated with cannabis-related variations in cortical thickness, and virtual histology revealed coexpression patterns of these 13 genes with cell-specific markers of astrocytes, microglia, and a type of pyramidal cells enriched in dendrite-regulating genes. Similarly, the spatial expression of 18 WIN-related human homologs of DEGs correlated with group differences in cortical thickness and showed coexpression patterns with the same three cell types. Gene ontology analysis indicated that 37 THC-related human homologs are enriched in neuron projection development, while 33 WIN-related homologs are enriched in processes associated with learning and memory. In mice, we observed spine loss and lower dendritic complexity in pyramidal cells of THC-exposed animals (vs controls). Experimentation with cannabis during adolescence may influence cortical thickness by impacting glutamatergic synapses and dendritic arborization.
Collapse
Affiliation(s)
- Xavier Navarri
- Department of Neuroscience, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
| | | | - Iness Charfi
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Florian Wünnemann
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | | | - Giacomo Trottier
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Sévérine Leclerc
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Gregor U Andelfinger
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Graziella Di Cristo
- Department of Neuroscience, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Louis Richer
- Department of Health Sciences, Université du Québec à Chicoutimi, Chicoutimi, Quebec G7H 2B1, Canada
| | - G Bruce Pike
- Departments of Radiology and Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Zdenka Pausova
- Departments of Physiology and Nutritional Sciences, Hospital for Sick Children, University of Toronto, Peter Gilgan Centre for Research and Learning, Toronto, Ontario M5G 0A4, Canada
| | - Graciela Piñeyro
- Department of Neuroscience, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Tomáš Paus
- Department of Neuroscience, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Psychiatry and Addictology, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| |
Collapse
|
3
|
Piscura MK, Henderson-Redmond AN, Barnes RC, Mitra S, Guindon J, Morgan DJ. Mechanisms of cannabinoid tolerance. Biochem Pharmacol 2023; 214:115665. [PMID: 37348821 PMCID: PMC10528043 DOI: 10.1016/j.bcp.2023.115665] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
Cannabis has been used recreationally and medically for centuries, yet research into understanding the mechanisms of its therapeutic effects has only recently garnered more attention. There is evidence to support the use of cannabinoids for the treatment of chronic pain, muscle spasticity, nausea and vomiting due to chemotherapy, improving weight gain in HIV-related cachexia, emesis, sleep disorders, managing symptoms in Tourette syndrome, and patient-reported muscle spasticity from multiple sclerosis. However, tolerance and the risk for cannabis use disorder are two significant disadvantages for cannabinoid-based therapies in humans. Recent work has revealed prominent sex differences in the acute response and tolerance to cannabinoids in both humans and animal models. This review will discuss evidence demonstrating cannabinoid tolerance in rodents, non-human primates, and humans and our current understanding of the neuroadaptations occurring at the cannabinoid type 1 receptor (CB1R) that are responsible tolerance. CB1R expression is downregulated in tolerant animals and humans while there is strong evidence of CB1R desensitization in cannabinoid tolerant rodent models. Throughout the review, critical knowledge gaps are indicated and discussed, such as the lack of a neuroimaging probe to assess CB1R desensitization in humans. The review discusses the intracellular signaling pathways that are responsible for mediating CB1R desensitization and downregulation including the action of G protein-coupled receptor kinases, β-arrestin2 recruitment, c-Jun N-terminal kinases, protein kinase A, and the intracellular trafficking of CB1R. Finally, the review discusses approaches to reduce cannabinoid tolerance in humans based on our current understanding of the neuroadaptations and mechanisms responsible for this process.
Collapse
Affiliation(s)
- Mary K Piscura
- Department of Biomedical Sciences, Marshall University, Huntington, WV 25755, USA; Department of Biomedical Sciences, Edward Via College of Osteopathic Medicine, Auburn, AL 36832, USA
| | | | - Robert C Barnes
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Swarup Mitra
- Department of Biomedical Sciences, Marshall University, Huntington, WV 25755, USA
| | - Josée Guindon
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Daniel J Morgan
- Department of Biomedical Sciences, Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|
4
|
Nealon CM, Henderson-Redmond AN, Hale DE, Morgan DJ. Tolerance to WIN55,212-2 is delayed in desensitization-resistant S426A/S430A mice. Neuropharmacology 2019; 148:151-159. [PMID: 30629988 PMCID: PMC6535342 DOI: 10.1016/j.neuropharm.2018.12.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 12/05/2018] [Accepted: 12/21/2018] [Indexed: 02/08/2023]
Abstract
Tolerance to cannabinoid agonists can develop through desensitization of the cannabinoid receptor 1 (CB1) following prolonged administration. Desensitization results from phosphorylation of CB1 by a G protein-coupled receptor kinase (GRK), and subsequent association of the receptor with arrestin. Mice expressing a mutant form of CB1, in which the serine residues at two putative phosphorylation sites necessary for desensitization have been replaced by non-phosphorylatable alanines (S426A/S430A), display reduced tolerance to Δ9-tetrahydrocannabinol (Δ9-THC). Tolerance to the antinociceptive effects of WIN55,212-2 was delayed in S426A/S430A mutants using the tail-flick and formalin tests. However, tolerance to the antinociceptive effects of once daily CP55,940 injections was not significantly delayed in S426A/S430A mutant mice using either of these tests. Interestingly, the dose response curve shifts for the hypothermic and antinociceptive effects of CP55,940 that were induced by chronic treatment with this agonist in wild-type mice were blocked in S426A/S430A mutant mice. Assessment of mechanical allodynia in mice exhibiting chronic cisplatin-evoked neuropathic pain found that tolerance to the anti-allodynic effects WIN55,212-2 but not CP55,940 was delayed in S426A/S430A mice compared to wild-type littermates. Despite these deficits in tolerance, S426A/S430A mutant mice eventually developed tolerance to both WIN55,212-2 and CP55,940 for all pain assays that were examined, suggesting that other mechanisms likely contribute to tolerance for these cannabinoid agonists. These findings suggest that GRK- and βarrestin2-mediated desensitization of CB1 may strongly contribute to the rate of tolerance to the antinociceptive effects of WIN55,212-2, and raises the possibility of agonist-specific mechanisms of cannabinoid tolerance.
Collapse
Affiliation(s)
- Caitlin M Nealon
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Angela N Henderson-Redmond
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - David E Hale
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Daniel J Morgan
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA; Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, 17033, USA; Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
5
|
Keenan CM, Storr MA, Thakur GA, Wood JT, Wager-Miller J, Straiker A, Eno MR, Nikas SP, Bashashati M, Hu H, Mackie K, Makriyannis A, Sharkey KA. AM841, a covalent cannabinoid ligand, powerfully slows gastrointestinal motility in normal and stressed mice in a peripherally restricted manner. Br J Pharmacol 2015; 172:2406-18. [PMID: 25572435 DOI: 10.1111/bph.13069] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/19/2014] [Accepted: 01/02/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Cannabinoid (CB) ligands have been demonstrated to have utility as novel therapeutic agents for the treatment of pain, metabolic conditions and gastrointestinal (GI) disorders. However, many of these ligands are centrally active, which limits their usefulness. Here, we examine a unique novel covalent CB receptor ligand, AM841, to assess its potential for use in physiological and pathophysiological in vivo studies. EXPERIMENTAL APPROACH The covalent nature of AM841 was determined in vitro using electrophysiological and receptor internalization studies on isolated cultured hippocampal neurons. Mouse models were used for behavioural analysis of analgesia, hypothermia and hypolocomotion. The motility of the small and large intestine was assessed in vivo under normal conditions and after acute stress. The brain penetration of AM841 was also determined. KEY RESULTS AM841 behaved as an irreversible CB1 receptor agonist in vitro. AM841 potently reduced GI motility through an action on CB1 receptors in the small and large intestine under physiological conditions. AM841 was even more potent under conditions of acute stress and was shown to normalize accelerated GI motility under these conditions. This compound behaved as a peripherally restricted ligand, showing very little brain penetration and no characteristic centrally mediated CB1 receptor-mediated effects (analgesia, hypothermia or hypolocomotion). CONCLUSIONS AND IMPLICATIONS AM841, a novel peripherally restricted covalent CB1 receptor ligand that was shown to be remarkably potent, represents a new class of potential therapeutic agents for the treatment of functional GI disorders.
Collapse
Affiliation(s)
- C M Keenan
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Alberta, Canada; Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Morphine mediates a proinflammatory phenotype via μ-opioid receptor–PKCɛ–Akt–ERK1/2 signaling pathway in activated microglial cells. Biochem Pharmacol 2013; 86:487-96. [DOI: 10.1016/j.bcp.2013.05.027] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/28/2013] [Accepted: 05/31/2013] [Indexed: 11/18/2022]
|
7
|
Selective kinase inhibitors as tools for neuroscience research. Neuropharmacology 2012; 63:1227-37. [DOI: 10.1016/j.neuropharm.2012.07.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 07/06/2012] [Accepted: 07/11/2012] [Indexed: 01/02/2023]
|
8
|
Zeng L, Webster SV, Newton PM. The biology of protein kinase C. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:639-61. [PMID: 22453963 DOI: 10.1007/978-94-007-2888-2_28] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This review gives a basic introduction to the biology of protein kinase C, one of the first calcium-dependent kinases to be discovered. We review the structure and function of protein kinase C, along with some of the substrates of individual isoforms. We then review strategies for inhibiting PKC in experimental systems and finally discuss the therapeutic potential of targeting PKC. Each aspect is covered in summary, with links to detailed resources where appropriate.
Collapse
Affiliation(s)
- Lily Zeng
- School of Medicine, University of California, San Francisco, CA, USA
| | | | | |
Collapse
|
9
|
Asimaki O, Mangoura D. Cannabinoid receptor 1 induces a biphasic ERK activation via multiprotein signaling complex formation of proximal kinases PKCε, Src, and Fyn in primary neurons. Neurochem Int 2010; 58:135-44. [PMID: 21074588 DOI: 10.1016/j.neuint.2010.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 11/02/2010] [Accepted: 11/03/2010] [Indexed: 11/17/2022]
Abstract
Cannabinoid receptors 1 (CB1Rs) play important roles in the regulation of dendritic branching, synapse density, and synaptic transmission through multiple G-protein-coupled signaling systems, including the activation of the extracellular signal-regulated kinases ERK1/2. The proximal signaling interactions leading to ERK1/2 activation by CB1R in CNS remain, however, unclear. Here, we present evidence that the CB1R agonist methanandamide induced a biphasic and sustained activation of ERK1/2 in primary neurons derived from E7 telencephalon. We show that E7 neurons natively express high levels of CB1R message and protein, the majority of which associates with PKCɛ at basal conditions. We now demonstrate that the first peak of ERK activation by CB1R was mediated by the sequential activation of G(q), PLC, and PKCɛ, selectively, and that the CB1R-activated PKCɛ acutely formed transient signaling modules containing activated Src and Fyn. A second pool of CB1Rs, coupled to PTX-sensitive activation of G(i/o), utilized as effectors additional Src and Fyn molecules to generate a second, additional wave of ERK activation at 15 min. Concurrently to these intermolecular signaling interactions, cytoskeleton-associated proteins MARCKS and p120catenin were drastically modified by phosphorylation of PKC and Src, respectively. These receptor-proximal signaling events correlated well with induction of neuritic outgrowth in the long term. Our data provide evidence for multiprotein signaling complex formation in the coupling of CB1R to activation of ERK in CNS neurons, and may elucidate several of the less understood acute effects of cannabinoid drugs.
Collapse
Affiliation(s)
- Olga Asimaki
- Developmental Neurobiology and Neurochemistry Group, Basic Neurosciences, Center for Preventive Medicine, Neurosciences and Social Psychiatry, Biomedical Research Foundation of the Academy of Athens, 4, Soranou Ephessiou Street, 11527 Athens, Greece
| | | |
Collapse
|
10
|
Olive MF, Newton PM. Protein kinase C isozymes as regulators of sensitivity to and self-administration of drugs of abuse-studies with genetically modified mice. Behav Pharmacol 2010; 21:493-9. [PMID: 20671547 PMCID: PMC3070663 DOI: 10.1097/fbp.0b013e32833d8bb7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Studies using targeted gene deletion in mice have revealed distinct roles for individual isozymes of the protein kinase C (PKC) family of enzymes in regulating sensitivity to various drugs of abuse. These changes in drug sensitivity are associated with altered patterns of drug self-administration. The purpose of this review is to summarize behavioral studies conducted on mice carrying targeted deletions of genes encoding specific PKC isozymes (namely the beta, gamma, delta, and epsilon isozymes), and to critically evaluate the possibility of using pharmacological inhibitors of specific PKC isozymes as modulators of the sensitivity to various drugs of abuse, as well as potential aids in the treatment of substance use disorders.
Collapse
Affiliation(s)
- Michael Foster Olive
- Department of Psychiatry, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | |
Collapse
|
11
|
Roberto M, Cruz M, Bajo M, Siggins GR, Parsons LH, Schweitzer P. The endocannabinoid system tonically regulates inhibitory transmission and depresses the effect of ethanol in central amygdala. Neuropsychopharmacology 2010; 35:1962-72. [PMID: 20463657 PMCID: PMC2904853 DOI: 10.1038/npp.2010.70] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 03/21/2010] [Accepted: 04/11/2010] [Indexed: 11/08/2022]
Abstract
The central amygdala (CeA) has a major role in alcohol dependence and reinforcement, and behavioral and neurochemical evidence suggests a role for the endocannabinoid (eCB) system in ethanol binging and dependence. We used a slice preparation to investigate the physiological role of cannabinoids and their interaction with ethanol on inhibitory synaptic transmission in CeA. Superfusion of the cannabinoid receptor (CB1) agonist WIN55212-2 (WIN2) onto CeA neurons decreased evoked GABA(A) receptor-mediated inhibitory postsynaptic potentials (IPSPs) in a concentration-dependent manner, an effect prevented by the CB1 antagonists Rimonabant (SR141716, SR1) and AM251. SR1 or AM251 applied alone augmented IPSPs, revealing a tonic eCB activity that decreased inhibitory transmission in CeA. Paired-pulse analysis suggested a presynaptic CB1 mechanism. Intracellular BAPTA abolished the ability of AM251 to augment IPSPs, demonstrating the eCB-driven nature and postsynaptic origin of the tonic CB1-dependent control of GABA release. Superfusion of ethanol increased IPSPs and addition of WIN2 reversed the ethanol effect. Similarly, previous superfusion of WIN2 prevented subsequent ethanol effects on GABAergic transmission. The ethanol-induced augmentation of IPSPs was additive to CB1 blockade, ruling out a participation of CB1 in the action of acute ethanol. Our study points to an important role of CB1 in CeA in which the eCBs tonically regulate neuronal activity, and suggests a potent mechanism for modulating CeA tone during challenge with ethanol.
Collapse
Affiliation(s)
- Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California, USA
- Pearson Center for Alcoholism and Addiction Research, The Scripps Research Institute, La Jolla, California, USA
| | - Maureen Cruz
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California, USA
| | - Michal Bajo
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California, USA
| | - George R Siggins
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California, USA
| | - Loren H Parsons
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California, USA
- Pearson Center for Alcoholism and Addiction Research, The Scripps Research Institute, La Jolla, California, USA
| | - Paul Schweitzer
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
12
|
Martini L, Thompson D, Kharazia V, Whistler JL. Differential regulation of behavioral tolerance to WIN55,212-2 by GASP1. Neuropsychopharmacology 2010; 35:1363-73. [PMID: 20164830 PMCID: PMC2953419 DOI: 10.1038/npp.2010.6] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 12/19/2009] [Accepted: 01/06/2010] [Indexed: 12/13/2022]
Abstract
Cannabinoid agonists have shown some promise clinically as analgesics, in particular for cancer pain, in which they have the additional benefit of decreasing nausea. However, as for most other drugs, the long-term use of cannabinoids is limited by the development of tolerance. Several molecular mechanisms have been proposed to explain drug tolerance, including receptor downregulation. The cannabinoid 1 (CB1) receptors can be downregulated in vitro through an interaction with the G-protein-coupled receptor-associated sorting protein1, GASP1, that targets CB1 receptors for degradation after their agonist-mediated endocytosis. To investigate whether GASP1-mediated postendocytic sorting of the CB1 receptor contributes to tolerance to cannabinoid drugs in vivo, we generated a mouse with a disruption of GASP1. In wild-type mice, repeated administration of the cannabinoid agonist WIN55,212-2 promoted downregulation of CB1 receptor levels and concomitant tolerance to the effects of drug on antinociception, motor incoordination, and locomotor hypoactivity. In contrast, GASP1 knockout mice did not develop tolerance to any of these effects and showed no significant receptor downregulation. Taken together, this study provides evidence that GASP1 regulates CB1 receptor downregulation in vivo, and that postendocytic receptor trafficking has a key role in the development of tolerance to WIN55,212-2.
Collapse
Affiliation(s)
- Lene Martini
- Ernest Gallo Clinic and Research Center, University of California San Francisco, Emeryville, CA, USA
| | - Dawn Thompson
- Ernest Gallo Clinic and Research Center, University of California San Francisco, Emeryville, CA, USA
| | - Viktor Kharazia
- Ernest Gallo Clinic and Research Center, University of California San Francisco, Emeryville, CA, USA
| | - Jennifer L Whistler
- Ernest Gallo Clinic and Research Center, University of California San Francisco, Emeryville, CA, USA
| |
Collapse
|
13
|
Järbe TUC, Li C, Vadivel SK, Makriyannis A. Discriminative stimulus functions of methanandamide and delta(9)-THC in rats: tests with aminoalkylindoles (WIN55,212-2 and AM678) and ethanol. Psychopharmacology (Berl) 2010; 208:87-98. [PMID: 19902182 PMCID: PMC3727230 DOI: 10.1007/s00213-009-1708-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Accepted: 10/22/2009] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The aim of the study was to characterize in vivo the aminoalkylindoles WIN55,212-2 (WIN) and AM678 (naphthalen-1-yl(1-pentyl-1H-indol-3-yl)methanone) as cannabinoid receptor (CB(1)R) ligands using drug discrimination. Tests also involved delta(9)-tetrahydrocannabinol (THC) and R-(+)-methanandamide (mAEA), a metabolically stable analog of the endogenous ligand anandamide, as well as the CB(1)R selective antagonist/inverse agonist rimonabant; tests with ethanol assessed pharmacological specificity. We used two different drug discriminations (mAEA and THC) allowing us to explore potential differences in CB(1)R activation which could be attributed to variations in their respective CB(1)R signaling mechanisms. METHODS There were two concurrently trained groups of rats. One group discriminated between i.p. injected vehicle and 10 mg/kg mAEA. The other group was trained to discriminate between vehicle and 1.8 mg/kg THC. RESULTS Dose generalization curves for AM678, WIN55,212-2, THC, and mAEA suggested the following rank order of potency: AM678 > WIN55,212-2 > or = THC > mAEA in both drug discrimination groups. Challenge by 1 mg/kg rimonabant resulted in shifts to the right of the generalization curves for the two aminoalkylindoles (4.4-fold for AM678 and 11.3-fold for WIN in the mAEA group, whereas for the THC group, the corresponding values were 13 and 2.6, respectively), suggesting surmountable antagonism. Ethanol did not generalize in either of the two groups, suggesting pharmacological specificity. CONCLUSION Data are congruent with the general observation that there is substantial overlap in the discriminative stimulus effects of CB(1)R ligands across different chemical classes. However, the quantitative differences in the interactions between the two aminoalkylindoles and rimonabant in the two discrimination groups suggest subtle variations in the ligand-receptor activation(s).
Collapse
Affiliation(s)
- Torbjörn U C Järbe
- Department of Psychology, Temple University, 265-67 Weiss Hall, 1701 North 13th Street, Philadelphia, PA 19122, USA.
| | | | | | | |
Collapse
|