1
|
Chakraborty S, Rao S, Tripathi SJ. The neuroprotective effects of N-acetylcysteine in psychiatric and neurodegenerative disorders: From modulation of glutamatergic transmission to restoration of synaptic plasticity. Neuropharmacology 2025:110527. [PMID: 40414419 DOI: 10.1016/j.neuropharm.2025.110527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 05/10/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
N-acetylcysteine (NAC) is an effective pleiotropic drug with a strong safety profile. It is predominantly used as a mucolytic agent and in the treatment of paracetamol overdose. However, extensive research in the last decade has shown the prominent efficacy of NAC in many neuropsychiatric and neurodegenerative disorders. NAC acts through multiple mechanisms; primarily, it releases cysteine and modulates glutamatergic and monoaminergic transmission. Further, it restores glutathione levels, promotes oxidative balance, reverses decreased synaptic plasticity, reduces neuroinflammation and mitochondrial dysfunction, and provides neurotrophic support. Additionally, it regulates one-carbon metabolism pathways, leading to the production of key metabolites. In this review, we will be discussing in-depth mechanisms of action of NAC and its promising ability to reverse neuropathological changes, particularly cognitive deficits, and associated plasticity changes in various psychiatric and neurodegenerative diseases, including depression, bipolar disorders, schizophrenia, Alzheimer's disease, Huntington's disease, traumatic brain injury, aging. Overall, several preclinical studies and clinical trials have demonstrated the efficacy of NAC in reversing regressive plasticity, cognitive deficits, and associated changes in the brain. NAC remains among the strongest candidates with a high safety profile for managing several types of neurological disorders.
Collapse
Affiliation(s)
- Suwarna Chakraborty
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shankaranarayana Rao
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India.
| | - Sunil Jamuna Tripathi
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
2
|
Lee S, Edwards S. Alcohol and cannabis use for pain management: Translational findings of relative risks, benefits, and interactions. Physiol Behav 2025; 294:114867. [PMID: 40023207 DOI: 10.1016/j.physbeh.2025.114867] [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: 10/15/2024] [Revised: 02/08/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Chronic pain affects over 20% of the global population and contributes to the vast burden of psychiatric illness. While effective treatments for chronic pain remain limited, both alcohol and cannabis have been used for centuries to manage pain and closely associated negative affective symptoms. However, persistent misuse of alcohol and/or cannabis in such a negative reinforcement fashion is hypothesized to increase the risk of severity of substance use disorders (SUDs). The current review describes neurobiological evidence for the analgesic efficacy of alcohol and primary cannabis constituents and how use or co-use of these substances may influence SUD risk.
Collapse
Affiliation(s)
- Sumin Lee
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, 2020 Gravier St. Room 734, New Orleans, LA 70112, USA
| | - Scott Edwards
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, 2020 Gravier St. Room 734, New Orleans, LA 70112, USA.
| |
Collapse
|
3
|
De Felice M, Szkudlarek HJ, Uzuneser TC, Rodríguez-Ruiz M, Sarikahya MH, Pusparajah M, Galindo Lazo JP, Whitehead SN, Yeung KKC, Rushlow WJ, Laviolette SR. The Impacts of Adolescent Cannabinoid Exposure on Striatal Anxiety- and Depressive-Like Pathophysiology Are Prevented by the Antioxidant N-Acetylcysteine. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100361. [PMID: 39257692 PMCID: PMC11381987 DOI: 10.1016/j.bpsgos.2024.100361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 09/12/2024] Open
Abstract
Background Exposure to Δ9-tetrahydrocannabinol (THC) is an established risk factor for later-life neuropsychiatric vulnerability, including mood- and anxiety-related symptoms. The psychotropic effects of THC on affect and anxiogenic behavioral phenomena are known to target the striatal network, particularly the nucleus accumbens, a neural region linked to mood and anxiety disorder pathophysiology. THC may increase neuroinflammatory responses via the redox system and dysregulate inhibitory and excitatory neural balance in various brain circuits, including the striatum. Thus, interventions that can induce antioxidant effects may counteract the neurodevelopmental impacts of THC exposure. Methods In the current study, we used an established preclinical adolescent rat model to examine the impacts of adolescent THC exposure on various behavioral, molecular, and neuronal biomarkers associated with increased mood and anxiety disorder vulnerability. Moreover, we investigated the protective properties of the antioxidant N-acetylcysteine against THC-related pathology. Results We demonstrated that adolescent THC exposure induced long-lasting anxiety- and depressive-like phenotypes concomitant with differential neuronal and molecular abnormalities in the two subregions of the nucleus accumbens, the shell and the core. In addition, we report for the first time that N-acetylcysteine can prevent THC-induced accumbal pathophysiology and associated behavioral abnormalities. Conclusions The preventive effects of this antioxidant intervention highlight the critical role of redox mechanisms underlying cannabinoid-induced neurodevelopmental pathology and identify a potential intervention strategy for the prevention and/or reversal of these pathophysiological sequelae.
Collapse
Affiliation(s)
- Marta De Felice
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Hanna J Szkudlarek
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Taygun C Uzuneser
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mar Rodríguez-Ruiz
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mohammed H Sarikahya
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | | | | | - Shawn N Whitehead
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Ken K-C Yeung
- Department of Chemistry, Western University, London, Ontario, Canada
- Department of Biochemistry, Western University, London, Ontario, Canada
| | - Walter J Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute (CHRI), London, Ontario, Canada
| |
Collapse
|
4
|
Corley C, Craig A, Sadek S, Marusich JA, Chehimi SN, White AM, Holdiness LJ, Reiner BC, Gipson CD. Enhancing translation: A need to leverage complex preclinical models of addictive drugs to accelerate substance use treatment options. Pharmacol Biochem Behav 2024; 243:173836. [PMID: 39067531 PMCID: PMC11344688 DOI: 10.1016/j.pbb.2024.173836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Preclinical models of addictive drugs have been developed for decades to model aspects of the clinical experience in substance use disorders (SUDs). These include passive exposure as well as volitional intake models across addictive drugs and have been utilized to also measure withdrawal symptomatology and potential neurobehavioral mechanisms underlying relapse to drug seeking or taking. There are a number of Food and Drug Administration (FDA)-approved medications for SUDs, however, many demonstrate low clinical efficacy as well as potential sex differences, and we also note gaps in the continuum of care for certain aspects of clinical experiences in individuals who use drugs. In this review, we provide a comprehensive update on both frequently utilized and novel behavioral models of addiction with a focus on translational value to the clinical experience and highlight the need for preclinical research to follow epidemiological trends in drug use patterns to stay abreast of clinical treatment needs. We then note areas in which models could be improved to enhance the medications development pipeline through efforts to enhance translation of preclinical models. Next, we describe neuroscience efforts that can be leveraged to identify novel biological mechanisms to enhance medications development efforts for SUDs, focusing specifically on advances in brain transcriptomics approaches that can provide comprehensive screening and identification of novel targets. Together, the confluence of this review demonstrates the need for careful selection of behavioral models and methodological parameters that better approximate the clinical experience combined with cutting edge neuroscience techniques to advance the medications development pipeline for SUDs.
Collapse
Affiliation(s)
- Christa Corley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ashley Craig
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Safiyah Sadek
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Samar N Chehimi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley M White
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Lexi J Holdiness
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
5
|
Oliva HNP, Prudente TP, Nunes EJ, Cosgrove KP, Radhakrishnan R, Potenza MN, Angarita GA. Substance use and spine density: a systematic review and meta-analysis of preclinical studies. Mol Psychiatry 2024; 29:2873-2885. [PMID: 38561468 DOI: 10.1038/s41380-024-02519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
The elucidation of synaptic density changes provides valuable insights into the underlying brain mechanisms of substance use. In preclinical studies, synaptic density markers, like spine density, are altered by substances of abuse (e.g., alcohol, amphetamine, cannabis, cocaine, opioids, nicotine). These changes could be linked to phenomena including behavioral sensitization and drug self-administration in rodents. However, studies have produced heterogeneous results for spine density across substances and brain regions. Identifying patterns will inform translational studies given tools that now exist to measure in vivo synaptic density in humans. We performed a meta-analysis of preclinical studies to identify consistent findings across studies. PubMed, ScienceDirect, Scopus, and EBSCO were searched between September 2022 and September 2023, based on a protocol (PROSPERO: CRD42022354006). We screened 6083 publications and included 70 for meta-analysis. The meta-analysis revealed drug-specific patterns in spine density changes. Hippocampal spine density increased after amphetamine. Amphetamine, cocaine, and nicotine increased spine density in the nucleus accumbens. Alcohol and amphetamine increased, and cannabis reduced, spine density in the prefrontal cortex. There was no convergence of findings for morphine's effects. The effects of cocaine on the prefrontal cortex presented contrasting results compared to human studies, warranting further investigation. Publication bias was small for alcohol or morphine and substantial for the other substances. Heterogeneity was moderate-to-high across all substances. Nonetheless, these findings inform current translational efforts examining spine density in humans with substance use disorders.
Collapse
Affiliation(s)
- Henrique Nunes Pereira Oliva
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA
| | - Tiago Paiva Prudente
- Faculdade de Medicina, Universidade Federal de Goiás (UFG), Goiânia, Goiás, Brazil
| | - Eric J Nunes
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Yale Tobacco Center of Regulatory Science, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly P Cosgrove
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Rajiv Radhakrishnan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Marc N Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
- Connecticut Council on Problem Gambling, Wethersfield, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Gustavo A Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA.
| |
Collapse
|
6
|
Freels TG, Westbrook SR, Zamberletti E, Kuyat JR, Wright HR, Malena AN, Melville MW, Brown AM, Glodosky NC, Ginder DE, Klappenbach CM, Delevich KM, Rubino T, McLaughlin RJ. Sex Differences in Response-Contingent Cannabis Vapor Administration During Adolescence Mediate Enduring Effects on Behavioral Flexibility and Prefrontal Microglia Activation in Rats. Cannabis Cannabinoid Res 2024; 9:e1184-e1196. [PMID: 38190273 PMCID: PMC11392456 DOI: 10.1089/can.2023.0014] [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] [Indexed: 01/10/2024] Open
Abstract
Introduction: Cannabis is the most used illicit drug in the United States. With many states passing legislation to permit its recreational use, there is concern that cannabis use among adolescents could increase dramatically in the coming years. Historically, it has been difficult to model real-world cannabis use to investigate the causal relationship between cannabis use in adolescence and behavioral and neurobiological effects in adulthood. Materials and Methods: We used a response-contingent vapor administration model to investigate long-term effects of cannabis use during adolescence on the medial prefrontal cortex (mPFC) and mPFC-dependent behaviors in male and female rats. Results: Adolescent (35- to 55-day-old) female rats had significantly higher rates of responding for vaporized Δ9-tetrahydrocannabinol (THC)-dominant cannabis extract (CANTHC) compared with adolescent males. In adulthood (70-110 days old), female, but not male, CANTHC rats also took more trials to reach criterion and made more regressive errors in an automated attentional set-shifting task compared with vehicle rats, thereby indicating sex differences in behavioral flexibility impairments. Notably, sex-treatment interactions were not observed when rats of each sex were exposed to a noncontingent CANTHC vapor dosing regimen that approximated CANTHC vapor deliveries earned by females. No differences were observed in effort-based decision making in either sex. In the mPFC, female (but not male) CANTHC rats displayed more reactive microglia with no changes in myelin basic protein expression or dendritic spine density. Conclusion: Altogether, these data reveal important sex differences in rates of responding for CANTHC vapor in adolescence that may confer enduring alterations to mPFC structure and function and suggest that there may be subtle differences in the effects of response-contingent versus noncontingent cannabis exposure that should be systematically examined in future studies.
Collapse
Affiliation(s)
- Timothy G. Freels
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Sara R. Westbrook
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Erica Zamberletti
- Department of Biotechnology and Life Sciences and Neuroscience Center, University of Insubria, Busto Arsizio, Varese, Italy
| | - Jacqulyn R. Kuyat
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Hayden R. Wright
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Alexandra N. Malena
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Max W. Melville
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Amanda M. Brown
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | | | - Darren E. Ginder
- Department of Psychology, Washington State University, Pullman, Washington, USA
| | - Courtney M. Klappenbach
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Kristen M. Delevich
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences and Neuroscience Center, University of Insubria, Busto Arsizio, Varese, Italy
| | - Ryan J. McLaughlin
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
- Department of Psychology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
7
|
Mustafa MA, Poklis JL, Karin KN, Elmer JA, Porter JH, Parra V, Lu D, Schlosburg JE, Lichtman AH. Investigation of Cannabidiol in the Mouse Drug Discrimination Paradigm. Cannabis Cannabinoid Res 2024; 9:581-590. [PMID: 36656312 PMCID: PMC10998012 DOI: 10.1089/can.2022.0198] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Introduction: Cannabidiol (CBD) has gained considerable public and scientific attention because of its known and potential medicinal properties, as well as its commercial success in a wide range of products. Although CBD lacks cannabimimetic intoxicating side effects in humans and fails to substitute for cannabinoid type-1 receptor (CB1R) agonists in laboratory animal models of drug discrimination paradigm, anecdotal reports describe it as producing a "pleasant" subjective effect in humans. Thus, we speculated that this phytocannabinoid may elicit distinct subjective effects. Accordingly, we investigated whether mice would learn to discriminate CBD from vehicle. Additionally, we examined whether CBD may act as a CB1R allosteric and whether it would elevate brain endocannabinoid concentrations. Materials and Methods: C57BL/6J mice underwent discrimination training of either CBD or the high-efficacy CB1R agonist CP55,940 from vehicle. Additionally, we examined whether CBD or the CB1R-positive allosteric modulator ZCZ011 would alter the CP55,940 discriminative cue. Finally, we tested whether an acute CBD injection would elevate endocannabinoid levels in brain, and also quantified blood and brain levels of CBD. Results: Mice failed to discriminate high doses of CBD from vehicle following 124 training days, though the same subjects subsequently acquired CP55,940 discrimination. In a second group of mice trained to discriminate CP55,940, CBD neither elicited substitution nor altered response rates. A single injection of 100 or 200 mg/kg CBD did not affect brain levels of endogenous cannabinoids and related lipids and resulted in high drug concentrations in blood and whole brain at 0.5 h and continued to increase at 3 h. Discussion: CBD did not engender an interoceptive stimulus, did not disrupt performance in a food-motivated operant task, and lacked apparent effectiveness in altering brain endocannabinoid levels or modulating the pharmacological effects of a CB1R agonist. These findings support the assertions that CBD lacks abuse liability and its acute administration does not appear to play a functional role in modulating key components of the endocannabinoid system in whole animals.
Collapse
Affiliation(s)
- Mohammed A. Mustafa
- Department of Pharmacology and Toxicology, and Virginia Commonwealth University, Richmond, Virginia, USA
| | - Justin L. Poklis
- Department of Pharmacology and Toxicology, and Virginia Commonwealth University, Richmond, Virginia, USA
| | - Kimberly N. Karin
- Department of Pharmacology and Toxicology, and Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jayden A. Elmer
- Department of Pharmacology and Toxicology, and Virginia Commonwealth University, Richmond, Virginia, USA
| | - Joseph H. Porter
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Victoria Parra
- Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas, USA
| | - Dai Lu
- Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas, USA
| | - Joel E. Schlosburg
- Department of Pharmacology and Toxicology, and Virginia Commonwealth University, Richmond, Virginia, USA
| | - Aron H. Lichtman
- Department of Pharmacology and Toxicology, and Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
8
|
Gutierrez A, Creehan KM, Grant Y, Taffe MA. Adult consequences of repeated nicotine and Δ 9-tetrahydrocannabinol (THC) vapor inhalation in adolescent rats. Psychopharmacology (Berl) 2024; 241:585-599. [PMID: 38282127 PMCID: PMC10884208 DOI: 10.1007/s00213-024-06545-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/20/2024] [Indexed: 01/30/2024]
Abstract
RATIONALE Use of electronic drug delivery systems (EDDS, "e-cigarettes") to ingest nicotine and Δ9-tetrahydrocannabinol (THC) has surged in adolescents in the USA; five times as many high-school seniors vape nicotine daily using tobacco. At the same time, 19.5% of seniors use cannabis at least monthly, with 12% using EDDS to deliver it. OBJECTIVES This study was conducted to examine the impact of repeated adolescent vapor inhalation of nicotine and THC in rats. METHODS Female Sprague-Dawley rats were exposed to 30-min sessions of vapor inhalation, twice daily, from post-natal day (PND) 31 to PND 40. Conditions included vapor from the propylene glycol (PG) vehicle, nicotine (60 mg/mL in the PG), THC (100 mg/mL in the PG), or the combination of nicotine (60 mg/mL) and THC (100 mg/mL). Rats were assessed on wheel activity, heroin anti-nociception and nicotine and heroin vapor volitional exposure during adulthood. RESULTS Nicotine-exposed rats exhibited few differences as adults, but were less sensitive to anti-nociceptive effects of heroin (1 mg/kg, s.c.). THC- and THC + nicotine-exposed rats were less spontaneously active, and obtained fewer nicotine vapor deliveries as adults. In contrast, THC-exposed rats obtained volitional heroin vapor at rates indistinguishable from the non-THC-exposed groups. Repeated THC exposure also caused tolerance to temperature-disrupting effects of THC (5 mg/kg, i.p.). CONCLUSIONS These studies further confirm that the effects of repeated vapor exposure to THC in adolescence last into early to middle adulthood, including decreased volitional consumption of nicotine. Effects of repeated nicotine in adolescence were comparatively minor.
Collapse
Affiliation(s)
- Arnold Gutierrez
- Department of Psychiatry, University of California, 9500 Gilman Drive, San Diego, La Jolla, CA, 92093, USA
| | - Kevin M Creehan
- Department of Psychiatry, University of California, 9500 Gilman Drive, San Diego, La Jolla, CA, 92093, USA
| | - Yanabel Grant
- Department of Psychiatry, University of California, 9500 Gilman Drive, San Diego, La Jolla, CA, 92093, USA
| | - Michael A Taffe
- Department of Psychiatry, University of California, 9500 Gilman Drive, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
9
|
Brewer AL, Felter CE, Sternitzky AR, Spencer SM. Somatic and anxiety-like behaviors in male and female rats during withdrawal from the non-selective cannabinoid agonist WIN 55,212-2. Pharmacol Biochem Behav 2024; 236:173707. [PMID: 38244864 PMCID: PMC10923112 DOI: 10.1016/j.pbb.2024.173707] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 01/22/2024]
Abstract
Synthetic cannabinoids are associated with higher risk of dependence and more intense withdrawal symptoms than plant-derived Δ9-tetrahydrocannabinol (THC). Avoidance of withdrawal symptoms, including anxiogenic effects, can contribute to continued cannabinoid use. Adult male and female Long-Evans rats were given escalating doses of WIN 55,212-2 (WIN) via twice daily intrajugular infusions. Precipitated withdrawal was elicited with SR 141716 (rimonabant) 4 h after the final infusion. Global withdrawal scores (GWS) were compiled by summing z-scores of observed somatic behaviors over a 30-min period with locomotor activity simultaneously collected via beam breaks. Rimonabant precipitated withdrawal in female and male rats at 3 or 10 mg/kg, respectively, but the individual behaviors contributing to GWS were not identical. 3 mg/kg rimonabant did not impact locomotor behavior in females, but 10 mg/kg decreased locomotion in male controls. Spontaneous withdrawal observed between 6 and 96 h after the final infusion was quantifiable up to 24 h following WIN administration. Individual behaviors contributing to GWS varied by sex and time point. Males undergoing spontaneous withdrawal engaged in more locomotion than females undergoing withdrawal. Separate groups of rats were subjected to a battery of anxiety-like behavioral tests (elevated plus maze, open field test, and marble burying test) one or two weeks after WIN or vehicle infusions. At one week abstinence, sex-related effects were noted in marble burying and the open field test but were unrelated to drug treatment. At two weeks abstinence, females undergoing withdrawal spent more time grooming during marble burying and performed more marble manipulations than their male counterparts. WIN infusions did not impact estrous cycling, and GWS scores were not correlated with estrous at withdrawal. Collectively, these results show qualitative sex differences in behaviors contributing to the behavioral experience of cannabinoid withdrawal supporting clinical findings from THC.
Collapse
Affiliation(s)
- Abigail L Brewer
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States of America; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, MN, United States of America.
| | - Claire E Felter
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States of America; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, MN, United States of America
| | - Anna R Sternitzky
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States of America; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, MN, United States of America
| | - Sade M Spencer
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States of America; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, MN, United States of America
| |
Collapse
|
10
|
Eckard ML, Kinsey SG. Differential disruption of response alternation by precipitated Δ 9-THC withdrawal and subsequent Δ 9-THC abstinence in mice. Pharmacol Biochem Behav 2024; 236:173718. [PMID: 38272272 PMCID: PMC10955601 DOI: 10.1016/j.pbb.2024.173718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/01/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
In addition to overt somatic symptoms, cannabinoid withdrawal can also manifest as disruptions in motivation and attention. Experimental animal models using operant-conditioning approaches reveal these differences, in either antagonist-precipitated or spontaneous withdrawal models. However, these processes have yet to be characterized in the same subjects simultaneously. To differentiate between motivational and attentional processes disrupted in cannabinoid withdrawal, the current study used a response alternation task in which a fixed-ratio (FR) schedule repeatedly alternated between two spatially distinct response options throughout daily training sessions. This task yielded traditional measures of motivation (e.g., response latency) as well as attention (e.g., responses to the incorrect side). After two weeks of training, male and female C57BL/6 J mice either received vehicle or Δ9-THC (10 mg/kg, s.c.) twice daily for 5 days. On the 6th day, all mice received their final injection of vehicle or Δ9-THC followed 30 min later by injection of the CB1 receptor selective inverse agonist rimonabant (2 mg/kg, i.p.) to precipitate withdrawal. Testing continued for 3 days post-rimonabant to assess how THC abstinence impacted task performance. Whereas rimonabant decreased response rates to equal degrees in THC-treated and vehicle-treated mice, THC-treated mice showed longer session times, longer response latencies, and more errors per reinforcer. Only THC-treated mice showed a longer latency to switch after committing an error reflecting that precipitated withdrawal impacted measures of both motivation and attention. During the 3-day abstinence window, performance of vehicle-treated mice returned to baseline, but THC-treated mice continued to show disruptions in motivational measures. Importantly, attentional measures (errors and latency to switch after an error) were unaffected by THC abstinence. These data suggest that precipitated and "spontaneous" cannabinoid withdrawal may be qualitatively and quantitatively distinct withdrawal conditions with precipitated withdrawal disrupting both attentional and motivational processes, while abstinence may only affect motivation.
Collapse
Affiliation(s)
| | - Steven G Kinsey
- School of Nursing, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
11
|
Liu J, Nabavizadeh P, Rao P, Derakhshandeh R, Han DD, Guo R, Murphy MB, Cheng J, Schick SF, Springer ML. Impairment of Endothelial Function by Aerosol From Marijuana Leaf Vaporizers. J Am Heart Assoc 2023; 12:e032969. [PMID: 38014661 PMCID: PMC10727338 DOI: 10.1161/jaha.123.032969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 11/02/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Marijuana leaf vaporizers, which heat plant material and sublimate Δ-9-tetrahydrocannabinol without combustion, are popular alternatives to smoking cannabis that are generally perceived to be less harmful. We have shown that smoke from tobacco and marijuana, as well as aerosol from e-cigarettes and heated tobacco products, impair vascular endothelial function in rats measured as arterial flow-mediated dilation (FMD). METHODS AND RESULTS We exposed 8 rats per group to aerosol generated by 2 vaporizer systems (Volcano and handheld Yocan) using marijuana with varying Δ-9-tetrahydrocannabinol levels, in a single pulsatile exposure session of 2 s/min over 5 minutes, and measured changes in FMD. To model secondhand exposure, we exposed rats for 1 minute to diluted aerosol approximating release of uninhaled Volcano aerosol into typical residential rooms. Exposure to aerosol from marijuana with and without cannabinoids impaired FMD by ≈50%. FMD was similarly impaired by aerosols from Yocan (237 °C), and from Volcano at both its standard temperature (185 °C) and the minimum sublimation temperature of Δ-9-tetrahydrocannabinol (157 °C), although the low-temperature aerosol condition did not effectively deliver Δ-9-tetrahydrocannabinol to the circulation. Modeled secondhand exposure based on diluted Volcano aerosol also impaired FMD. FMD was not affected in rats exposed to clean air or water vapor passed through the Volcano system. CONCLUSIONS Acute direct exposure and modeled secondhand exposure to marijuana leaf vaporizer aerosol, regardless of cannabinoid concentration or aerosol generation temperature, impair endothelial function in rats comparably to marijuana smoke. Our findings indicate that use of leaf vaporizers is unlikely to reduce the vascular risk burden of smoking marijuana.
Collapse
Affiliation(s)
- Jiangtao Liu
- Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoCAUSA
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Pooneh Nabavizadeh
- Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoCAUSA
- Present address:
Division of CardiologyUniversity of CincinnatiCincinnatiOHUSA
| | - Poonam Rao
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCAUSA
- Center for Tobacco Control Research and EducationUniversity of California, San FranciscoSan FranciscoCAUSA
- Present address:
Christus Good Shepherd/Texas A&M University Internal Medicine Residency ProgramLongviewTXUSA
| | - Ronak Derakhshandeh
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Daniel D. Han
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCAUSA
- Present address:
School of Medicine and DentistryUniversity of RochesterRochesterNYUSA
| | - Raymond Guo
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCAUSA
- Present address:
Stanford UniversityStanfordCAUSA
| | - Morgan B. Murphy
- Division of Occupational and Environmental MedicineUniversity of California, San FranciscoSan FranciscoCAUSA
- Present address:
Sutter Health California Pacific Medical CenterStanfordCAUSA
| | - Jing Cheng
- Division of Oral Epidemiology and Dental Public HealthUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Suzaynn F. Schick
- Center for Tobacco Control Research and EducationUniversity of California, San FranciscoSan FranciscoCAUSA
- Division of Occupational and Environmental MedicineUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Matthew L. Springer
- Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoCAUSA
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCAUSA
- Center for Tobacco Control Research and EducationUniversity of California, San FranciscoSan FranciscoCAUSA
| |
Collapse
|
12
|
Abraham AD, Wiley JL, Marusich JA. Experimenter administered Δ 9-THC decreases nicotine self-administration in a rat model. Pharmacol Biochem Behav 2023; 231:173632. [PMID: 37690617 PMCID: PMC10543614 DOI: 10.1016/j.pbb.2023.173632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/22/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND The co-use of nicotine and cannabis has been steadily rising in the United States. Rodent studies suggest that delta-9-tetrahydrocannabinol (THC) could increase addictive qualities of nicotine, but whether repeated THC exposure alters self-administration of nicotine has not been tested. We hypothesized that THC would increase the reinforcing effects of nicotine and alter nicotine intake. METHODS Adult male and female Sprague-Dawley rats were treated with THC (0, 3, 30 mg/kg) daily for 14 days prior to and during training for intravenous self-administration of nicotine. Rats were allowed to self-administer nicotine for several weeks, then tested for sensitivity to nicotine dose through multiple determinations of a nicotine dose-effect curve with or without THC pretreatment. A separate set of rats were trained on fixed ratio responding for sucrose and assessed for THC effects on behavior. RESULTS Post-session THC decreased nicotine self-administration in male and female rats throughout acquisition and maintenance and increased the latency to stable rates of nicotine intake during acquisition. Post-session THC shifted nicotine dose-effect curves downward, and pre-session THC suppressed responding at higher nicotine doses. Unlike nicotine, responding for sucrose was not affected by post-session THC. Pre-session THC decreased responding for sucrose, particularly for THC-naïve rats. CONCLUSIONS Repeated post-session THC decreased nicotine-taking behaviors but did not alter sucrose responding. Thus, post-session THC may alter sensitivity to nicotine. Pre-session THC treatment decreased lever pressing in both sucrose and nicotine studies, indicating this effect was nonspecific. These studies show that THC modulates patterns of nicotine intake in rat models.
Collapse
Affiliation(s)
- Antony D Abraham
- Center for Drug Discovery, RTI International, 3040 Cornwallis Rd, Research Triangle Park, NC 27709, USA
| | - Jenny L Wiley
- Center for Drug Discovery, RTI International, 3040 Cornwallis Rd, Research Triangle Park, NC 27709, USA
| | - Julie A Marusich
- Center for Drug Discovery, RTI International, 3040 Cornwallis Rd, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
13
|
Gutierrez A, Creehan KM, Grant Y, Taffe MA. Adult consequences of repeated nicotine and Δ 9 -tetrahydrocannabinol (THC) vapor inhalation in adolescent rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.08.556932. [PMID: 37745433 PMCID: PMC10515744 DOI: 10.1101/2023.09.08.556932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The use of Electronic Drug Delivery Systems (EDDS, "e-cigarettes") to ingest nicotine and Δ 9 -tetrahydrocannabinol (THC) has surged in adolescent populations in the United States, as five times as many high-school seniors vape nicotine daily as use tobacco. At the same time 19.5% of seniors use cannabis at least monthly, with 12% using EDDS to deliver it. This study was conducted to examine the impact of repeated adolescent vapor inhalation of nicotine and THC in rats. Female Sprague-Dawley rats were exposed to 30-minute sessions of vapor inhalation, twice daily, from Post-Natal Day (PND) 31 to PND 40. Conditions included vapor from the propylene glycol (PG) vehicle, Nicotine (60 mg/mL in the PG), THC (100 mg/mL in the PG) or the combination of Nicotine (60 mg/mL) and THC (100 mg/mL). Rats were assessed on wheel activity, heroin anti-nociception and nicotine and heroin vapor volitional exposure during adulthood. Nicotine exposed rats exhibited few differences as adults, but were less sensitive to anti-nociceptive effects of heroin (1 mg/kg, s.c.). THC- and THC+Nicotine-exposed rats were less spontaneously active, and obtained fewer nicotine vapor deliveries as adults. In contrast, THC exposed rats obtained volitional heroin vapor at rates indistinguishable from the non-THC-exposed groups. Repeated THC exposure also caused tolerance to temperature-disrupting effects of THC (5 mg/kg, i.p.). These studies further confirm that the effects of repeated vapor exposure to THC in adolescence last into early to middle adulthood, including decreased volitional consumption of nicotine. Effects of repeated nicotine in adolescence were comparatively minor.
Collapse
|
14
|
Garcia-Keller C, Hohmeister M, Seidling K, Beloate L, Chioma V, Spencer S, Kalivas P, Neuhofer D. Δ 9 -Tetrahydrocannabinol self-administration induces cell type-specific adaptations in the nucleus accumbens core. Addict Biol 2023; 28:e13286. [PMID: 37500492 PMCID: PMC10924663 DOI: 10.1111/adb.13286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 07/29/2023]
Abstract
Drugs of abuse induce cell type-specific adaptations in D1- and D2-medium spiny neurons (MSNs) in the nucleus accumbens core (NAcore) that can bias signalling towards D1-MSNs and enhance relapse vulnerability. Whether Δ9 -tetrahydrocannabinol (THC) use initiates similar neuroadaptations is unknown. D1- and D2-Cre transgenic rats were transfected with Cre-dependent reporters and trained to self-administer THC + cannabidiol (THC + CBD). After extinction training spine morphology, glutamate transmission, CB1R function and cFOS expression were quantified. We found that extinction from THC + CBD induced a loss of large spine heads in D1- but not D2-MSNs and commensurate reductions in glutamate synaptic transmission. Also, presynaptic CB1R function was impaired selectively at glutamatergic synapses on D1-MSNs, which augmented the capacity to potentiate glutamate transmission. Using cFOS expression as an activity marker, we found no change after extinction but increased cFOS expression in D1-MSNs after cue-induced drug seeking. Contrasting D1-MSNs, CB1R function and glutamate synaptic transmission on D2-MSN synapses were unaffected by THC + CBD use. However, cFOS expression was decreased in D2-MSNs of THC + CBD-extinguished rats and was restored after drug seeking. Thus, CB1R adaptations in D1-MSNs partially predicted neuronal activity changes, posing pathway specific modulation of eCB signalling in D1-MSNs as a potential treatment avenue for cannabis use disorder (CUD).
Collapse
Affiliation(s)
- Constanza Garcia-Keller
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226
| | - Madeline Hohmeister
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| | - Kailyn Seidling
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| | - Lauren Beloate
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| | - Vivian Chioma
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| | - Sade Spencer
- Department of Pharmacology, Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, MN 55455
| | - Peter Kalivas
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| | - Daniela Neuhofer
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| |
Collapse
|
15
|
Herbst ED, Pennington DL, Borsari B, Manuel J, Yalch M, Alcid E, Martinez Rivas M, Delacruz J, Rossi N, Garcia B, Wong N, Batki SL. N-acetylcysteine for smoking cessation among dual users of tobacco and cannabis: Protocol and rationale for a randomized controlled trial. Contemp Clin Trials 2023; 131:107250. [PMID: 37271412 PMCID: PMC10847050 DOI: 10.1016/j.cct.2023.107250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND Tobacco and cannabis co-use is a growing public health problem. The synergistic effects of cannabis and nicotine on neurobiological systems that mediate reward and shared environmental cues reinforcing use may make tobacco smoking cessation more difficult. N-acetylcysteine (NAC), an FDA-approved medication and over-the-counter supplement, has shown promise in animal studies and randomized controlled trials (RCTs) in reducing tobacco and cannabis craving and use. NAC's potential efficacy in treating addiction may be attributable to its central nervous system effects in reducing excessive glutamatergic activity, oxidative stress, and inflammation. To date, no RCT has examined NAC for smoking cessation among dual users of tobacco and cannabis. METHOD In a double-blind, placebo-controlled RCT, we will examine NAC for smoking cessation among dual users of tobacco and cannabis. Sixty adult cigarette-cannabis co-users are randomized to receive NAC 3600 mg per day or placebo over 8 weeks. Participants in both groups receive 8 weekly cognitive behavioral therapy sessions addressing smoking cessation and cannabis reduction. Outcomes are assessed at Weeks 0, 4, 8, and 12. Primary aims are to determine NAC's efficacy in decreasing cigarette craving, nicotine dependence, and use; and cannabis craving and use. Exploratory aims include examination of changes in neurocognition with NAC and their potential mediational effects on cigarette and cannabis use outcomes. CONCLUSION Results will inform smoking cessation treatment among dual users of tobacco and cannabis. CLINICALTRIALS gov Identifier: NCT04627922.
Collapse
Affiliation(s)
- Ellen D Herbst
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA.
| | - David L Pennington
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Brian Borsari
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Jennifer Manuel
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Matthew Yalch
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA; Palo Alto University, 1791 Arastradero Rd., Palo Alto, CA 94304, USA
| | - Eric Alcid
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Madeline Martinez Rivas
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Joannalyn Delacruz
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Nathan Rossi
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Brianna Garcia
- California Institute of Technology, 1200 E California Blvd, Pasadena, CA 91125, USA
| | - Natalie Wong
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Steven L Batki
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| |
Collapse
|
16
|
Kalivas PW, Gourley SL, Paulus MP. Intrusive thinking: Circuit and synaptic mechanisms of a transdiagnostic psychiatric symptom. Neurosci Biobehav Rev 2023; 150:105196. [PMID: 37094741 PMCID: PMC10249786 DOI: 10.1016/j.neubiorev.2023.105196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 04/26/2023]
Abstract
Spontaneous thought is an adaptive cognitive process that can produce novel and insightful thought sequences useful in guiding future behavior. In many psychiatric disorders, spontaneous thinking becomes intrusive and uncontrolled, and can trigger symptoms such as craving, repetitive negative thinking and trauma-related memories. We link studies using clinical imaging and rodent modeling towards understanding the neurocircuitry and neuroplasticity of intrusive thinking. We propose a framework in which drugs or stress change the homeostatic set point of brain reward circuitry, which then impacts subsequent plasticity induced by drug/stress conditioned cues (metaplastic allostasis). We further argue for the importance of examining not only the canonical pre- and postsynapse, but also the adjacent astroglial protrusions and extracellular matrix that together form the tetrapartite synapse and that plasticity throughout the tetrapartite synapse is necessary for cue-induced drug or stress behaviors. This analysis reveals that drug use or trauma cause long-lasting allostatic brain plasticity that sets the stage for subsequent drug/trauma-associated cues to induce transient plasticity that can lead to intrusive thinking.
Collapse
Affiliation(s)
- Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| | - Shannon L Gourley
- Emory National Primate Research Center, Emory University, Department of Pediatrics and Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA; Emory National Primate Research Center, Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|
17
|
Lewandowski SI, Hodebourg R, Wood SK, Carter JS, Nelson KH, Kalivas PW, Reichel CM. Matrix metalloproteinase activity during methamphetamine cued relapse. Addict Biol 2023; 28:e13279. [PMID: 37186441 PMCID: PMC10506177 DOI: 10.1111/adb.13279] [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: 11/08/2022] [Revised: 01/23/2023] [Accepted: 03/27/2023] [Indexed: 05/17/2023]
Abstract
Relapse to drug seeking involves transient synaptic remodelling that occurs in response to drug-associated cues. This remodelling includes activation of matrix metalloproteinases (MMPs) to initiate catalytic signalling in the extracellular matrix in the nucleus accumbens core (NAcore). We hypothesized that MMP activity would be increased in the NAcore during cue-induced methamphetamine (meth) seeking in a rat model of meth use and relapse. Male and female rats had indwelling jugular catheters and bilateral intracranial cannula targeting the NAcore surgically implanted. Following recovery, rats underwent meth or saline self-administration (6 h/day for 15 days) in which active lever responding was paired with a light + tone stimulus complex, followed by home cage abstinence. Testing occurred after 7 or 30 days of abstinence. On test day, rats were microinjected with a fluorescein isothiocyanate (FITC)-quenched gelatin substrate that fluoresces following cleavage by MMP-2,9, allowing for the quantification of gelatinase activity during cued-relapse testing. MMP-2,9 activity was significantly increased in the NAcore by meth cues presentation after 7 and 30 days of abstinence, indicating that remodelling by MMPs occurs during presentation of meth associated cues. Surprisingly, although cue-induced seeking increased between Days 7 and 30, MMP-2,9 activity did not increase. These findings indicate that although MMP activation is elicited during meth cue-induced seeking, MMP activation did not parallel the meth seeking that occurs during extended drug abstinence.
Collapse
Affiliation(s)
- Stacia I. Lewandowski
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Ritchy Hodebourg
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Samuel K. Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Jordan S. Carter
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Katherine H. Nelson
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Carmela M. Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
18
|
Stringfield SJ, Sanders BE, Suppo JA, Sved AF, Torregrossa MM. Nicotine Enhances Intravenous Self-administration of Cannabinoids in Adult Rats. Nicotine Tob Res 2023; 25:1022-1029. [PMID: 36426873 PMCID: PMC10077937 DOI: 10.1093/ntr/ntac267] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/24/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Nicotine and cannabis are commonly used together, yet few studies have investigated the effects of concurrent administration. Nicotine exhibits reinforcement enhancing effects by promoting the reinforcing properties of stimuli including other drugs. As many studies of this effect used non-contingent nicotine, we implemented a dual-self-administration model where rats have simultaneous access to two drugs and choose which to self-administer throughout a session. Here, we investigated the effect of self-administered or non-contingently delivered nicotine on cannabinoid self-administration. METHODS Adult male rats were allowed to self-administer the synthetic cannabinoid WIN 55,212-2 (WIN) intravenously, with or without subcutaneous nicotine injections before each session. A separate group of animals were allowed to self-administer WIN, nicotine, or saline using a dual-catheter procedure, where each solution was infused independently and associated with a separate operant response. A third group of male and female rats were allowed to self-administer delta-9-tetrahydrocannabinol (THC) with or without pre-session injections of nicotine. RESULTS Nicotine injections increased self-administration of WIN and THC. During dual self-administration, nicotine availability increased saline and WIN infusions but nicotine intake was not changed by WIN or saline availability. Rats preferred nicotine over saline, but preferred nicotine and WIN equally when both were available. The effect of nicotine on cannabinoid self-administration was acute and reversible when nicotine was no longer present. CONCLUSIONS These results expand our understanding of the ability of nicotine to enhance reinforcement of other drugs and suggest that co-use of nicotine and cannabinoids promotes cannabinoid use beyond what would be taken alone. IMPLICATIONS This study utilizes a dual intravenous self-administration model to investigate the ability of nicotine to enhance cannabinoid intake. Our results demonstrate that the reinforcement enhancing properties of nicotine on drug use extend to include cannabinoids, but that this effect occurs specifically when nicotine is administered alongside the cannabinoid. Interestingly, cannabinoid use did not promote nicotine intake, suggesting this mechanism of reinforcement is specific to nicotine.
Collapse
Affiliation(s)
| | - Bryson E Sanders
- Department of Neuroscience, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jude A Suppo
- Department of Neuroscience, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alan F Sved
- Department of Neuroscience, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mary M Torregrossa
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
19
|
Cajiao-Manrique MDM, Maldonado R, Martín-García E. A male mouse model of WIN 55,212-2 self-administration to study cannabinoid addiction. Front Pharmacol 2023; 14:1143365. [PMID: 37050910 PMCID: PMC10083303 DOI: 10.3389/fphar.2023.1143365] [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: 01/12/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
We have established for the first time a mouse model of cannabinoid addiction using WIN 55,212-2 intravenous self-administration (0.0125 mg/kg/infusion) in C57Bl/6J mice. This model allows to evaluate the addiction criteria by grouping them into 1) persistence of response during a period of non-availability of the drug, 2) motivation for WIN 55,212-2 with a progressive ratio, and 3) compulsivity when the reward is associated with a punishment such as an electric foot-shock, in agreement with the Diagnostic and Statistical Manual of Mental Disorders 5th edition (DSM-5). This model also allows to measure two parameters that have been related with the DSM-5 diagnostic criteria of craving, resistance to extinction and reinstatement, and two phenotypic traits suggested as predisposing factors, impulsivity and sensitivity to reward. We found that 35.6% of mice developed the criteria of cannabinoid addiction, allowing to differentiate between resilient and vulnerable mice. Therefore, we have established a novel and reliable model to study the neurobiological correlates underlying the resilience or vulnerability to develop cannabinoid addiction. This model included the chemogenetic inhibition of neuronal activity in the medial prefrontal cortex to the nucleus accumbens pathway to assess the neurobiological substrate of cannabinoid addiction. This model will shed light on the neurobiological substrate underlying cannabinoid addiction.
Collapse
Affiliation(s)
- María del Mar Cajiao-Manrique
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Elena Martín-García
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
20
|
Buzzi B, Koseli E, Moncayo L, Shoaib M, Damaj M. Role of Neuronal Nicotinic Acetylcholine Receptors in Cannabinoid Dependence. Pharmacol Res 2023; 191:106746. [PMID: 37001709 DOI: 10.1016/j.phrs.2023.106746] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/15/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Cannabis is among the most widely consumed psychoactive drugs around the world and cannabis use disorder (CUD) has no current approved pharmacological treatment. Nicotine and cannabis are commonly co-used which suggests there to be overlapping neurobiological actions supported primarily by the co-distribution of both receptor systems in the brain. There appears to be strong rationale to explore the role that nicotinic receptors play in cannabinoid dependence. Preclinical studies suggest that the ɑ7 nAChR subtype may play a role in modulating the reinforcing and discriminative stimulus effects of cannabinoids, while the ɑ4β2 * nAChR subtype may be involved in modulating the motor and sedative effects of cannabinoids. Preclinical and human genetic studies point towards a potential role of the ɑ5, ɑ3, and β4 nAChR subunits in CUD, while human GWAS studies strongly implicate the ɑ2 subunit as playing a role in CUD susceptibility. Clinical studies suggest that current smoking cessation agents, such as varenicline and bupropion, may also be beneficial in treating CUD, although more controlled studies are necessary. Additional behavioral, molecular, and mechanistic studies investigating the role of nAChR in the modulation of the pharmacological effects of cannabinoids are needed.
Collapse
|
21
|
Freels TG, Westbrook SR, Wright HR, Kuyat JR, Zamberletti E, Malena AM, Melville MW, Brown AM, Glodosky NC, Ginder DE, Klappenbach CM, Delevich KM, Rubino T, McLaughlin RJ. Sex differences in adolescent cannabis vapor self-administration mediate enduring effects on behavioral flexibility and prefrontal microglia activation in rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.21.524468. [PMID: 36711651 PMCID: PMC9882275 DOI: 10.1101/2023.01.21.524468] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cannabis is the most used illicit drug in the United States. With many states passing legislation to permit its recreational use, there is concern that cannabis use among adolescents could increase dramatically in the coming years. Historically, it has been difficult to model real-world cannabis use to investigate the causal relationship between cannabis use in adolescence and behavioral and neurobiological effects in adulthood. To this end, we used a novel volitional vapor administration model to investigate long-term effects of cannabis use during adolescence on the medial prefrontal cortex (mPFC) and mPFC-dependent behaviors in male and female rats. Adolescent (35-55 day old) female rats had significantly higher rates of responding for vaporized Δ9-tetrahydrocannabinol (THC)-dominant cannabis extract (CANTHC) compared to adolescent males. In adulthood (70-110 day old), female, but not male, CANTHC rats also took more trials to reach criterion and made more regressive errors in an automated attentional set-shifting task compared to vehicle rats. Similar set-shifting deficits were observed in males when they were exposed to a non-contingent CANTHC vapor dosing regimen that approximated CANTHC self-administration rates in females. No differences were observed in effort-based decision making in either sex. In the mPFC, female (but not male) CANTHC rats displayed more reactive microglia with no significant changes in myelin basic protein expression or dendritic spine density. Together, these data reveal important sex differences in rates of cannabis vapor self-administration in adolescence that confer enduring alterations to mPFC structure and function. Importantly, female-specific deficits in behavioral flexibility appear to be driven by elevated rates of CANTHC self-administration as opposed to a sex difference in the effects of CANTHC vapor per se.
Collapse
Affiliation(s)
- Timothy G. Freels
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Sara R. Westbrook
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Hayden R. Wright
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Jacqulyn R. Kuyat
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Erica Zamberletti
- Department of Biotechnology and Life Sciences and Neuroscience Center, University of Insubria, Busto Arsizio, Varese, Italy
| | - Alexandra M. Malena
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Max W. Melville
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Amanda M. Brown
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | | | - Darren E. Ginder
- Department of Psychology, Washington State University, Pullman, WA, USA
| | - Courtney M. Klappenbach
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Kristen M. Delevich
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences and Neuroscience Center, University of Insubria, Busto Arsizio, Varese, Italy
| | - Ryan J. McLaughlin
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
- Department of Psychology, Washington State University, Pullman, WA, USA
| |
Collapse
|
22
|
Torrens A, Ruiz CM, Martinez MX, Tagne AM, Roy P, Grimes D, Ahmed F, Lallai V, Inshishian V, Bautista M, Chen YC, Huestis MA, Das A, Fowler CD, Mahler SV, Piomelli D. Nasal accumulation and metabolism of Δ 9-tetrahydrocannabinol following aerosol ('vaping') administration in an adolescent rat model. Pharmacol Res 2023; 187:106600. [PMID: 36481259 PMCID: PMC9845136 DOI: 10.1016/j.phrs.2022.106600] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Passive aerosol exposure to Δ9-tetrahydrocannabinol (THC) in laboratory animals results in faster onset of action and less extensive liver metabolism compared to most other administration routes and might thus provide an ecologically relevant model of human cannabis inhalation. Previous studies have, however, overlooked the possibility that rodents, as obligate nose breathers, may accumulate aerosolized THC in the nasal cavity, from where the drug might directly diffuse to the brain. To test this, we administered THC (ten 5-s puffs of 100 mg/mL of THC) to adolescent (31-day-old) Sprague-Dawley rats of both sexes. We used liquid chromatography/tandem mass spectrometry to quantify the drug and its first-pass metabolites - 11-hydroxy-Δ9-THC (11-OH-THC) and 11-nor-9-carboxy-Δ9-THC (11-COOH-THC) - in nasal mucosa, lungs, plasma, and brain (olfactory bulb and cerebellum) at various time points after exposure. Apparent maximal THC concentration and area under the curve were ∼5 times higher in nasal mucosa than in lungs and 50-80 times higher than in plasma. Concentrations of 11-OH-THC were also greater in nasal mucosa and lungs than other tissues, whereas 11-COOH-THC was consistently undetectable. Experiments with microsomal preparations confirmed local metabolism of THC into 11-OH-THC (not 11-COOH-THC) in nasal mucosa and lungs. Finally, whole-body exposure to THC deposited substantial amounts of THC (∼150 mg/g) on fur but suppressed post-exposure grooming in rats of both sexes. The results indicate that THC absorption and metabolism in nasal mucosa and lungs, but probably not gastrointestinal tract, contribute to the pharmacological effects of aerosolized THC in male and female rats.
Collapse
Affiliation(s)
- Alexa Torrens
- Department and Anatomy and Neurobiology, University of California, Irvine, CA 92697-4625, USA
| | - Christina M Ruiz
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4625, USA
| | - Maricela X Martinez
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4625, USA
| | - Alex Mabou Tagne
- Department and Anatomy and Neurobiology, University of California, Irvine, CA 92697-4625, USA
| | - Pritam Roy
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Dakota Grimes
- Department and Anatomy and Neurobiology, University of California, Irvine, CA 92697-4625, USA
| | - Faizy Ahmed
- Department and Anatomy and Neurobiology, University of California, Irvine, CA 92697-4625, USA
| | - Valeria Lallai
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4625, USA
| | - Victoria Inshishian
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4625, USA
| | - Malia Bautista
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4625, USA
| | - Yen-Chu Chen
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4625, USA
| | | | - Aditi Das
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Christie D Fowler
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4625, USA
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4625, USA
| | - Daniele Piomelli
- Department and Anatomy and Neurobiology, University of California, Irvine, CA 92697-4625, USA; Department of Biological Chemistry, University of California, Irvine, CA 92697-4625, USA; Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697-4625, USA.
| |
Collapse
|
23
|
Neutral CB1 Receptor Antagonists as Pharmacotherapies for Substance Use Disorders: Rationale, Evidence, and Challenge. Cells 2022; 11:cells11203262. [PMID: 36291128 PMCID: PMC9600259 DOI: 10.3390/cells11203262] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
Cannabinoid receptor 1 (CB1R) has been one of the major targets in medication development for treating substance use disorders (SUDs). Early studies indicated that rimonabant, a selective CB1R antagonist with an inverse agonist profile, was highly promising as a therapeutic for SUDs. However, its adverse side effects, such as depression and suicidality, led to its withdrawal from clinical trials worldwide in 2008. Consequently, much research interest shifted to developing neutral CB1R antagonists based on the recognition that rimonabant’s side effects may be related to its inverse agonist profile. In this article, we first review rimonabant’s research background as a potential pharmacotherapy for SUDs. Then, we discuss the possible mechanisms underlying its therapeutic anti-addictive effects versus its adverse effects. Lastly, we discuss the rationale for developing neutral CB1R antagonists as potential treatments for SUDs, the supporting evidence in recent research, and the challenges of this strategy. We conclude that developing neutral CB1R antagonists without inverse agonist profile may represent attractive strategies for the treatment of SUDs.
Collapse
|
24
|
Mechanistic Effects and Use of N-acetylcysteine in Substance Use Disorders. Curr Behav Neurosci Rep 2022. [DOI: 10.1007/s40473-022-00250-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
25
|
Therapeutic potential of PIMSR, a novel CB1 receptor neutral antagonist, for cocaine use disorder: evidence from preclinical research. Transl Psychiatry 2022; 12:286. [PMID: 35851573 PMCID: PMC9293959 DOI: 10.1038/s41398-022-02059-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/21/2022] [Accepted: 07/04/2022] [Indexed: 11/08/2022] Open
Abstract
Cannabinoid CB1 receptors (CB1Rs) have been major targets in medication development for the treatment of substance use disorders. However, clinical trials with rimonabant, a CB1R antagonist/inverse agonist, failed due to severe side effects. Here, we evaluated the therapeutic potential of PIMSR, a neutral CB1R antagonist lacking an inverse agonist profile, against cocaine's behavioral effects in experimental animals. We found that systemic administration of PIMSR dose-dependently inhibited cocaine self-administration under fixed-ratio (FR5), but not FR1, reinforcement, shifted the cocaine self-administration dose-response curve downward, decreased incentive motivation to seek cocaine under progressive-ratio reinforcement, and reduced cue-induced reinstatement of cocaine seeking. PIMSR also inhibited oral sucrose self-administration. Importantly, PIMSR alone is neither rewarding nor aversive as assessed by place conditioning. We then used intracranial self-stimulation (ICSS) to explore the possible involvement of the mesolimbic dopamine system in PIMSR's action. We found that PIMSR dose-dependently attenuated cocaine-enhanced ICSS maintained by electrical stimulation of the medial forebrain bundle in rats. PIMSR itself failed to alter electrical ICSS, but dose-dependently inhibited ICSS maintained by optical stimulation of midbrain dopamine neurons in transgenic DAT-Cre mice, suggesting the involvement of dopamine-dependent mechanisms. Lastly, we examined the CB1R mechanisms underlying PIMSR's action. We found that PIMSR pretreatment attenuated Δ9-tetrahydrocannabinol (Δ9-THC)- or ACEA (a selective CB1R agonist)-induced reduction in optical ICSS. Together, our findings suggest that the neutral CB1R antagonist PIMSR deserves further research as a promising pharmacotherapeutic for cocaine use disorder.
Collapse
|
26
|
Hasbi A, Madras BK, George SR. Daily THC and withdrawal increase dopamine D1-D2 receptor heteromer to mediate anhedonia and anxiogenic-like behavior through a dynorphin and kappa opioid receptor mechanism. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022. [PMID: 37519471 PMCID: PMC10382712 DOI: 10.1016/j.bpsgos.2022.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
Abstract
Background Frequent cannabis use is associated with a higher risk of developing cannabis use disorder and other adverse consequences. However, rodent models studying the underlying mechanisms of the reinforcing and withdrawal effects of the primary constituent of cannabis, Δ9-tetrahydrocannabinol (THC), have been limited. Methods This study investigated the effects of daily THC (1 mg/kg, intraperitoneal, 9 days) and spontaneous withdrawal (7 days) on hedonic and aversion-like behaviors in male rats. In parallel, underlying neuroadaptive changes in dopaminergic, opioidergic, and cannabinoid signaling in the nucleus accumbens were evaluated, along with a candidate peptide designed to reverse altered signaling. Results Chronic THC administration induced anhedonic- and anxiogenic-like behaviors not attributable to altered locomotor activity. These effects persisted after drug cessation. In the nucleus accumbens, THC treatment and withdrawal catalyzed increased cannabinoid CB1 receptor activity without modifying receptor expression. Dopamine D1-D2 receptor heteromer expression rose steeply with THC, accompanied by increased calcium-linked signaling, activation of BDNF/TrkB (brain-derived neurotrophic factor/tropomyosin receptor kinase B) pathway, dynorphin expression, and kappa opioid receptor signaling. Disruption of the D1-D2 heteromer by an interfering peptide during withdrawal reversed the anxiogenic-like and anhedonic-like behaviors as well as the neurochemical changes. Conclusions Chronic THC increases nucleus accumbens dopamine D1-D2 receptor heteromer expression and function, which results in increased dynorphin expression and kappa opioid receptor activation. These changes plausibly reduce dopamine release to trigger anxiogenic- and anhedonic-like behaviors after daily THC administration that persist for at least 7 days after drug cessation. These findings conceivably provide a therapeutic strategy to alleviate negative symptoms associated with cannabis use and withdrawal.
Collapse
|
27
|
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
|
28
|
Hodebourg R, Meyerink ME, Crow AD, Reichel CM, Kalivas PW, Garcia-Keller C. Cannabinoid use is enhanced by stress and changes conditioned stress responses. Neuropsychopharmacology 2022; 47:1037-1045. [PMID: 35145212 PMCID: PMC8938410 DOI: 10.1038/s41386-022-01287-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/16/2022] [Accepted: 01/26/2022] [Indexed: 11/08/2022]
Abstract
Individuals diagnosed with post-traumatic stress disorder (PTSD) are often comorbid for substance use disorders. Cannabis is widely used by PSTD patients, and the literature is mixed on whether cannabis use ameliorates or exacerbates patient responses to stress-associated conditioned stimuli (stress-CS). We determined if cannabis use affects responsivity to stress-CS in rats receiving 2 h stress in the presence of an odor stress-CS. Three weeks after acute stress, rats self-administered cannabinoids (delta9-tetrahydrocannabinol + cannabidiol; THC + CBD) for 15 days, and the stressed males consumed more THC + CBD than sham males. We then used the stress-CS or a novel odor (stress-NS) to reinstate THC + CBD seeking. Surprisingly, the stress-NS reinstated THC + CBD seeking, an effect blocked by N-acetylcysteine. Moreover, the stress-CS inhibited THC + CBD-CS induced reinstatement. To determine if the unexpected effects of stress-NS and -CS resulted from THC + CBD altering conditioned stress, the effect of THC + CBD use on stress-NS/CS-induced coping behaviors and spine morphology was quantified. In THC + CBD-treated rats, stress-NS increased active coping (burying). Conversely, stress-CS reduced active coping and increased passive coping (immobility) and other behavioral parameters associated with stress responses, including self-grooming and defecation. Transient spine head expansion in nucleus accumbens core is necessary for cue-induced drug seeking, and THC + CBD self-administration prevented the increase in head diameter by stress-CS in control rats. These data show THC + CBD self-administration altered the salience of environmental cues, causing neutral cues to promote active behavior (drug seeking and burying) and stress-CS to switch from active to passive behavior (inhibiting drug seeking and immobilization). We hypothesize that cannabis may exacerbate conditioned stress responses.
Collapse
Affiliation(s)
- Ritchy Hodebourg
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Michael E Meyerink
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Ayteria D Crow
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Carmela M Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| | | |
Collapse
|
29
|
Siemsen BM, Barry SM, Vollmer KM, Green LM, Brock AG, Westphal AM, King RA, DeVries DM, Otis JM, Cowan CW, Scofield MD. A Subset of Nucleus Accumbens Neurons Receiving Dense and Functional Prelimbic Cortical Input Are Required for Cocaine Seeking. Front Cell Neurosci 2022; 16:844243. [PMID: 35281297 PMCID: PMC8907444 DOI: 10.3389/fncel.2022.844243] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/02/2022] [Indexed: 11/24/2022] Open
Abstract
Background Prelimbic cortical projections to the nucleus accumbens core are critical for cue-induced cocaine seeking, but the identity of the accumbens neuron(s) targeted by this projection, and the transient neuroadaptations contributing to relapse within these cells, remain unknown. Methods Male Sprague-Dawley rats underwent cocaine or sucrose self-administration, extinction, and cue-induced reinstatement. Pathway-specific chemogenetics, patch-clamp electrophysiology, in vivo electrochemistry, and high-resolution confocal microscopy were used to identify and characterize a small population of nucleus accumbens core neurons that receive dense prelimbic cortical input to determine their role in regulating cue-induced cocaine and natural reward seeking. Results Chemogenetic inhibition of prelimbic cortical projections to the nucleus accumbens core suppressed cue-induced cocaine relapse and normalized real-time cue-evoked increases in accumbens glutamate release to that of sucrose seeking animals. Furthermore, chemogenetic inhibition of the population of nucleus accumbens core neurons receiving the densest prelimbic cortical input suppressed cocaine, but not sucrose seeking. These neurons also underwent morphological plasticity during the peak of cocaine seeking in the form of dendritic spine expansion and increased ensheathment by astroglial processes at large spines. Conclusion We identified and characterized a unique subpopulation of nucleus accumbens neurons that receive dense prelimbic cortical input. The functional specificity of this subpopulation is underscored by their ability to mediate cue-induced cocaine relapse, but not sucrose seeking. This subset of cells represents a novel target for addiction therapeutics revealed by anterograde targeting to interrogate functional circuits imbedded within a known network.
Collapse
Affiliation(s)
- Benjamin M. Siemsen
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Sarah M. Barry
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Kelsey M. Vollmer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Lisa M. Green
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Ashley G. Brock
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Annaka M. Westphal
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Raven A. King
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Derek M. DeVries
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - James M. Otis
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Christopher W. Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Michael D. Scofield
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, United States
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
30
|
Hodebourg R, Kalivas PW, Kruyer A. Extrasynaptic therapeutic targets in substance use and stress disorders. Trends Pharmacol Sci 2022; 43:56-68. [PMID: 34753604 PMCID: PMC8688303 DOI: 10.1016/j.tips.2021.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 01/03/2023]
Abstract
Treatments for substance use and stress disorders are based on ameliorating behavioral symptoms, not on reversing the synaptic pathology that has the potential to cure disorders. This failing arises in part from a research focus on how pre- and postsynaptic physiology is changed even though key neuropathology exists in the perisynaptic neuropil that homeostatically regulates synaptic transmission. We explore recent findings from the substance use and stress disorder literature pointing to a key role for perisynaptic astroglia and signaling in the extracellular matrix (ECM) in regulating synaptic pathology. We conclude that drugs and stress initiate long-lasting changes in brain synapses via enduring neuroadaptations in astroglia and the ECM, and that modulating extrasynaptic regulators may be therapeutically useful.
Collapse
Affiliation(s)
- Ritchy Hodebourg
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29464, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29464, USA.
| | - Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29464, USA
| |
Collapse
|
31
|
Bradlow RCJ, Berk M, Kalivas PW, Back SE, Kanaan RA. The Potential of N-Acetyl-L-Cysteine (NAC) in the Treatment of Psychiatric Disorders. CNS Drugs 2022; 36:451-482. [PMID: 35316513 PMCID: PMC9095537 DOI: 10.1007/s40263-022-00907-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/17/2022] [Indexed: 12/22/2022]
Abstract
N-acetyl-L-cysteine (NAC) is a compound of increasing interest in the treatment of psychiatric disorders. Primarily through its antioxidant, anti-inflammatory, and glutamate modulation activity, NAC has been investigated in the treatment of neurodevelopmental disorders, schizophrenia spectrum disorders, bipolar-related disorders, depressive disorders, anxiety disorders, obsessive compulsive-related disorders, substance-use disorders, neurocognitive disorders, and chronic pain. Whilst there is ample preclinical evidence and theoretical justification for the use of NAC in the treatment of multiple psychiatric disorders, clinical trials in most disorders have yielded mixed results. However, most studies have been underpowered and perhaps too brief, with some evidence of benefit only after months of treatment with NAC. Currently NAC has the most evidence of having a beneficial effect as an adjuvant agent in the negative symptoms of schizophrenia, severe autism, depression, and obsessive compulsive and related disorders. Future research with well-powered studies that are of sufficient length will be critical to better understand the utility of NAC in the treatment of psychiatric disorders.
Collapse
Affiliation(s)
| | - Michael Berk
- IMPACT-The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, VIC Australia ,Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Melbourne, VIC Australia ,Florey Institute of Neuroscience and Mental Health, Melbourne, VIC Australia ,Department of Psychiatry, University of Melbourne, Parkville, VIC Australia
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC USA ,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC USA
| | - Sudie E. Back
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC USA ,Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC USA
| | - Richard A. Kanaan
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC Australia ,Department of Psychiatry, University of Melbourne, Parkville, VIC Australia
| |
Collapse
|
32
|
Hempel B, Xi ZX. Receptor mechanisms underlying the CNS effects of cannabinoids: CB 1 receptor and beyond. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:275-333. [PMID: 35341569 PMCID: PMC10709991 DOI: 10.1016/bs.apha.2021.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Cannabis legalization continues to progress in many US states and other countries. Δ9-tetrahydrocannabinol (Δ9-THC) is the major psychoactive constituent in cannabis underlying both its abuse potential and the majority of therapeutic applications. However, the neural mechanisms underlying cannabis action are not fully understood. In this chapter, we first review recent progress in cannabinoid receptor research, and then examine the acute CNS effects of Δ9-THC or other cannabinoids (WIN55212-2) with a focus on their receptor mechanisms. In experimental animals, Δ9-THC or WIN55212-2 produces classical pharmacological effects (analgesia, catalepsy, hypothermia, hypolocomotion), biphasic changes in affect (reward vs. aversion, anxiety vs. anxiety relief), and cognitive deficits (spatial learning and memory, short-term memory). Accumulating evidence indicates that activation of CB1Rs underlies the majority of Δ9-THC or WIN55121-2's pharmacological and behavioral effects. Unexpectedly, glutamatergic CB1Rs preferentially underlie cannabis action relative to GABAergic CB1Rs. Functional roles for CB1Rs expressed on astrocytes and mitochondria have also been uncovered. In addition, Δ9-THC or WIN55212-2 is an agonist at CB2R, GPR55 and PPARγ receptors and recent studies implicate these receptors in a number of their CNS effects. Other receptors (such as serotonin, opioid, and adenosine receptors) also modulate Δ9-THC's actions and their contributions are detailed. This chapter describes the neural mechanisms underlying cannabis action, which may lead to new discoveries in cannabis-based medication development for the treatment of cannabis use disorder and other human diseases.
Collapse
Affiliation(s)
- Briana Hempel
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, United States.
| |
Collapse
|
33
|
Smiley CE, Saleh HK, Nimchuk KE, Garcia-Keller C, Gass JT. Adolescent exposure to delta-9-tetrahydrocannabinol and ethanol heightens sensitivity to fear stimuli. Behav Brain Res 2021; 415:113517. [PMID: 34389427 PMCID: PMC8404161 DOI: 10.1016/j.bbr.2021.113517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 01/05/2023]
Abstract
Cannabis use disorder (CUD) has doubled in prevalence over the past decade as a nation-wide trend toward legalization allows for increased drug accessibility. As a result, marijuana has become the most commonly used illicit drug in the United States particularly among the adolescent population. This is especially concerning since there is greater risk for the harmful side effects of drug use during this developmental period due to ongoing brain maturation. Increasing evidence indicates that CUD often occurs along with other debilitating conditions including both alcohol use disorder (AUD) and anxiety disorders such post-traumatic stress disorder (PTSD). Additionally, exposure to cannabis, alcohol, and stress can induce alterations in glutamate regulation and homeostasis in the prefrontal cortex (PFC) that may lead to impairments in neuronal functioning and cognition. Therefore, in order to study the relationship between drug exposure and the development of PTSD, these studies utilized rodent models to determine the impact of adolescent exposure to delta-9-tetrahydrocannabinol (THC) and ethanol on responses to fear stimuli during fear conditioning and used calcium imaging to measure glutamate activity in the prelimbic cortex during this behavioral paradigm. The results from these experiments indicate that adolescent exposure to THC and ethanol leads to enhanced sensitivity to fear stimuli both behaviorally and neuronally. Additionally, these effects were attenuated when animals were treated with the glutamatergic modulator N-acetylcysteine (NAC). In summary, these studies support the hypothesis that adolescent exposure to THC and ethanol leads to alterations in fear stimuli processing through glutamatergic reliant modifications in PFC signaling.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, United States.
| | - Heyam K Saleh
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, United States
| | - Katherine E Nimchuk
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, United States
| | - Constanza Garcia-Keller
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, United States
| | - Justin T Gass
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, United States
| |
Collapse
|
34
|
Finn DP, Haroutounian S, Hohmann AG, Krane E, Soliman N, Rice ASC. Cannabinoids, the endocannabinoid system, and pain: a review of preclinical studies. Pain 2021; 162:S5-S25. [PMID: 33729211 PMCID: PMC8819673 DOI: 10.1097/j.pain.0000000000002268] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/10/2021] [Indexed: 12/13/2022]
Abstract
ABSTRACT This narrative review represents an output from the International Association for the Study of Pain's global task force on the use of cannabis, cannabinoids, and cannabis-based medicines for pain management, informed by our companion systematic review and meta-analysis of preclinical studies in this area. Our aims in this review are (1) to describe the value of studying cannabinoids and endogenous cannabinoid (endocannabinoid) system modulators in preclinical/animal models of pain; (2) to discuss both pain-related efficacy and additional pain-relevant effects (adverse and beneficial) of cannabinoids and endocannabinoid system modulators as they pertain to animal models of pathological or injury-related persistent pain; and (3) to identify important directions for future research. In service of these goals, this review (1) provides an overview of the endocannabinoid system and the pharmacology of cannabinoids and endocannabinoid system modulators, with specific relevance to animal models of pathological or injury-related persistent pain; (2) describes pharmacokinetics of cannabinoids in rodents and humans; and (3) highlights differences and discrepancies between preclinical and clinical studies in this area. Preclinical (rodent) models have advanced our understanding of the underlying sites and mechanisms of action of cannabinoids and the endocannabinoid system in suppressing nociceptive signaling and behaviors. We conclude that substantial evidence from animal models supports the contention that cannabinoids and endocannabinoid system modulators hold considerable promise for analgesic drug development, although the challenge of translating this knowledge into clinically useful medicines is not to be underestimated.
Collapse
Affiliation(s)
- David P Finn
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, University Road, Galway, Ireland
| | - Simon Haroutounian
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Andrea G Hohmann
- Psychological and Brain Sciences, Program in Neuroscience, and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - Elliot Krane
- Departments of Anesthesiology, Perioperative, and Pain Medicine, & Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Nadia Soliman
- Pain Research, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, UK
| | - Andrew SC Rice
- Pain Research, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, UK
| |
Collapse
|
35
|
N-acetylcysteine in substance use disorder: a lesson from preclinical and clinical research. Pharmacol Rep 2021; 73:1205-1219. [PMID: 34091880 PMCID: PMC8460563 DOI: 10.1007/s43440-021-00283-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/19/2021] [Accepted: 05/19/2021] [Indexed: 12/29/2022]
Abstract
Substance use disorder (SUD) is a chronic brain condition, with compulsive and uncontrollable drug-seeking that leads to long-lasting and harmful consequences. The factors contributing to the development of SUD, as well as its treatment settings, are not fully understood. Alterations in brain glutamate homeostasis in humans and animals implicate a key role of this neurotransmitter in SUD, while the modulation of glutamate transporters has been pointed as a new strategy to diminish the excitatory glutamatergic transmission observed after drugs of abuse. N-acetylcysteine (NAC), known as a safe mucolytic agent, is involved in the regulation of this system and may be taken into account as a novel pharmacotherapy for SUD. In this paper, we summarize the current knowledge on the ability of NAC to reduce drug-seeking behavior induced by psychostimulants, opioids, cannabinoids, nicotine, and alcohol in animals and humans. Preclinical studies showed a beneficial effect in animal models of SUD, while the clinical efficacy of NAC has not been fully established. In summary, NAC will be a small add-on to usual treatment and/or psychotherapy for SUD, however, further studies are required.
Collapse
|
36
|
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: 27] [Impact Index Per Article: 6.8] [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
|
37
|
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: 1.0] [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
|
38
|
Hurel I, Muguruza C, Redon B, Marsicano G, Chaouloff F. Cannabis and exercise: Effects of Δ 9-tetrahydrocannabinol on preference and motivation for wheel-running in mice. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110117. [PMID: 32971218 DOI: 10.1016/j.pnpbp.2020.110117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/11/2020] [Accepted: 09/20/2020] [Indexed: 11/27/2022]
Abstract
Recent surveys have revealed close links between cannabis and exercise. Specifically, cannabis usage before and/or after exercise is an increasingly common habit primarily aimed at boosting exercise pleasure, motivation, and performance whilst facilitating post-exercise recovery. However, whether these beliefs reflect the true impact of cannabis on these aspects of exercise is unknown. This study has thus examined the effects of cannabis' main psychoactive ingredient, namely Δ9-tetrahydrocannabinol (THC), on (i) mouse wheel-running preference and performance and (ii) running motivation and seeking behaviour. Wheel-running preference and performance were investigated using a T-maze with free and locked wheels located at the extremity of either arm. Running motivation and seeking were assessed by a cued-running operant task wherein wheel-running was conditioned by nose poking. Moreover, because THC targets cannabinoid type 1 (CB1) receptors, i.e. receptors previously documented to control running motivation, this study also assessed the role of these receptors in running preference, performance, and craving-like behaviour. Whilst acute blockade or genetic deletion of CB1 receptors decreased running preference and performance in the T-maze, THC proved ineffective on either variable. The failure of THC to affect running variables in the T-maze extended to running motivation, as assessed by cued-running under a progressive ratio (PR) reinforcement schedule. This ineffectiveness of THC was not related to the treatment protocol because it successfully increased motivation for palatable food. Although craving-like behaviour, as indexed by a cue-induced reinstatement of running seeking, was found to depend on CB1 receptors, THC again proved ineffective. Neither running motivation nor running seeking were affected when CB1 receptors were further stimulated by increasing the levels of the endocannabinoid 2-arachidonoylglycerol. These results, which suggest that the drive for running is insensitive to the acute stimulation of CB1 receptors, raise the hypothesis that cannabis is devoid of effect on exercise motivation. Future investigation using chronic administration of THC, with and without other cannabis ingredients (e.g. cannabidiol), is however required before conclusions can be drawn.
Collapse
Affiliation(s)
- Imane Hurel
- Endocannabinoids and NeuroAdaptation, NeuroCentre INSERM U1215, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France
| | - Carolina Muguruza
- Endocannabinoids and NeuroAdaptation, NeuroCentre INSERM U1215, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France; Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain
| | - Bastien Redon
- Endocannabinoids and NeuroAdaptation, NeuroCentre INSERM U1215, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France
| | - Giovanni Marsicano
- Endocannabinoids and NeuroAdaptation, NeuroCentre INSERM U1215, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France
| | - Francis Chaouloff
- Endocannabinoids and NeuroAdaptation, NeuroCentre INSERM U1215, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France.
| |
Collapse
|
39
|
Effects of vapourized THC and voluntary alcohol drinking during adolescence on cognition, reward, and anxiety-like behaviours in rats. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110141. [PMID: 33069816 DOI: 10.1016/j.pnpbp.2020.110141] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/22/2020] [Accepted: 10/10/2020] [Indexed: 12/20/2022]
Abstract
Cannabis and alcohol co-use is prevalent in adolescence, but the long-term behavioural effects of this co-use remain largely unexplored. The aim of this study is to investigate the effects of adolescent alcohol and Δ9-tetrahydracannabinol (THC) vapour co-exposure on cognitive- and reward-related behaviours. Male Sprague-Dawley rats received vapourized THC (10 mg vapourized THC/four adolescent rats) or vehicle every other day (from post-natal day (PND) 28-42) and had continuous voluntary access to ethanol (10% volume/volume) in adolescence. Alcohol intake was measured during the exposure period to assess the acute effects of THC on alcohol consumption. In adulthood (PND 56+), rats underwent behavioural testing. Adolescent rats showed higher alcohol preference, assessed using the two-bottle choice test, on days on which they were not exposed to THC vapour. In adulthood, rats that drank alcohol as adolescents exhibited short-term memory deficits and showed decreased alcohol preference; on the other hand, rats exposed to THC vapour showed learning impairments in the delay-discounting task. Vapourized THC, alcohol or their combination had no effect on anxiety-like behaviours in adulthood. Our results show that although adolescent THC exposure acutely affects alcohol drinking, adolescent alcohol and cannabis co-use may not produce long-term additive effects.
Collapse
|
40
|
Martín-Sánchez A, García-Baos A, Castro-Zavala A, Alegre-Zurano L, Valverde O. Early-life stress exacerbates the effects of WIN55,212-2 and modulates the cannabinoid receptor type 1 expression. Neuropharmacology 2021; 184:108416. [PMID: 33271186 DOI: 10.1016/j.neuropharm.2020.108416] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 01/06/2023]
Abstract
Early-life stress induces an abnormal brain development and increases the risk of psychiatric diseases, including depression, anxiety and substance use disorders. We have developed a reliable model for maternal neglect, named maternal separation with early weaning (MSEW) in CD1 mice. In the present study, we evaluated the long-term effects on anxiety-like behaviours, nociception as well as the Iba1-positive microglial cells in this model in comparison to standard nest (SN) mice. Moreover, we investigated whether MSEW alters the cannabinoid agonist WIN55,212-2 effects regarding reward, spatial and emotional memories, tolerance to different cannabinoid responses, and physical dependence. Adult male offspring of MSEW group showed impaired responses on spatial and emotional memories after a repeated WIN55,212-2 treatment. These behavioural impairments were associated with an increase in basolateral amygdala and hippocampal CB1-expressing fibres and higher number of CB1-containing cells in cerebellum. Additionally, MSEW promotes a higher number of Iba1-positive microglial cells in basolateral amygdala and cerebellum. As for the cannabinoid-induced effects, rearing conditions did not influence the rewarding effects of WIN55,212-2 in the conditioned place preference paradigm. However, MSEW mice showed a delay in the development of tolerance to the cannabinoid effects. Moreover, CB1-positive fibres were reduced in limbic areas in MSEW mice after cannabinoid withdrawal precipitated with the CB1 antagonist SR141617A. These findings support that early-life stress promotes behavioural and molecular changes in the sensitivity to cannabinoids, which are mediated by alterations in CB1 signalling in limbic areas and it induces an increased Iba1-microglial marker which could interfere in emotional memories formation.
Collapse
Affiliation(s)
- Ana Martín-Sánchez
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Program, IMIM-Hospital Del Mar Research Institute, Barcelona, Spain
| | - Alba García-Baos
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Adriana Castro-Zavala
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laia Alegre-Zurano
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Program, IMIM-Hospital Del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
41
|
Gomez DM, Everett TJ, Hamilton LR, Ranganath A, Cheer JF, Oleson EB. Chronic cannabinoid exposure produces tolerance to the dopamine releasing effects of WIN 55,212-2 and heroin in adult male rats. Neuropharmacology 2021; 182:108374. [PMID: 33115642 PMCID: PMC7836093 DOI: 10.1016/j.neuropharm.2020.108374] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/16/2020] [Accepted: 10/24/2020] [Indexed: 02/06/2023]
Abstract
Synthetic cannabinoids were introduced into recreational drug culture in 2008 and quickly became one of the most commonly abused drugs in the United States. The neurobiological consequences resulting from synthetic cannabinoid repeated exposure remain poorly understood. It is possible that a blunted dopamine (DA) response may lead drug users to consume larger quantities to compensate for this form of neurochemical tolerance. Because the endogenous cannabinoid and opioid systems exhibit considerable cross-talk and cross-tolerance frequently develops following repeated exposure to either opioids or cannabinoids, there is interest in investigating whether a history of synthetic cannabinoid exposure influences the ability of heroin to increase DA release. To test the effects of chronic cannabinoid exposure on cannabinoid- and heroin-evoked DA release, male adult rats were treated with either vehicle or a synthetic cannabinoid (WIN55-212-2; WIN) using an intravenous (IV) dose escalation regimen (0.2-0.8 mg/kg IV over 9 treatments). As predicted, WIN-treated rats showed a rightward shift in the dose-response relationship across all behavioral/physiological measures when compared to vehicle-treated controls. Then, using fast-scan cyclic voltammetry to measure changes in the frequency of transient DA events in the nucleus accumbens shell of awake and freely-moving rats, it was observed that the DA releasing effects of both WIN and heroin were significantly reduced in male rats with a pharmacological history of cannabinoid exposure. These results demonstrate that repeated exposure to the synthetic cannabinoid WIN can produce tolerance to its DA releasing effects and cross-tolerance to the DA releasing effects of heroin.
Collapse
Affiliation(s)
- Devan M Gomez
- Psychology Department, University of Colorado Denver, USA; Current: Department of Biomedical Sciences, Marquette University, USA
| | | | | | - Ajit Ranganath
- Department of Neurobiology and Anatomy, University of Maryland Baltimore, USA
| | - Joseph F Cheer
- Department of Neurobiology and Anatomy, University of Maryland Baltimore, USA
| | - Erik B Oleson
- Psychology Department, University of Colorado Denver, USA; Biology Department, University of Colorado Denver, USA.
| |
Collapse
|
42
|
Stringfield SJ, Torregrossa MM. Intravenous self-administration of delta-9-THC in adolescent rats produces long-lasting alterations in behavior and receptor protein expression. Psychopharmacology (Berl) 2021; 238:305-319. [PMID: 33111197 PMCID: PMC7796919 DOI: 10.1007/s00213-020-05684-9] [Citation(s) in RCA: 16] [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: 04/22/2020] [Accepted: 10/19/2020] [Indexed: 12/18/2022]
Abstract
RATIONALE Initial exposure to cannabinoids, including Δ-9-tetrahydrocannabinol (THC), often occurs during adolescence. Considerable neurodevelopmental alterations occur throughout adolescence, and the environmental insult posed by exogenous cannabinoid exposure may alter natural developmental trajectories. Multiple studies suggest that long-lasting deficits in cognitive function occur as a result of adolescent cannabis use, but considerable variability exists in the magnitude of these effects. OBJECTIVES We sought to establish a novel procedure for achieving intravenous THC self-administration in adolescent rats in order to determine if volitional THC intake in adolescence produced indices of addiction-related behavior, altered working memory performance in adulthood, or altered the expression of proteins associated with these behaviors across several brain regions. METHODS Male and female adolescent rats learned to operantly self-administer escalating doses of THC intravenously from PD 32-51. Upon reaching adulthood they were tested in abstinence for cued reinstatement of THC-seeking and working memory performance on a delayed-match-to-sample task. In a separate cohort, glutamatergic, GABAergic, and cannabinoid receptor protein expression was measured in multiple brain regions. RESULTS Both male and female adolescents self-administered THC and exhibited cue-induced lever pressing throughout abstinence. THC-exposed males exhibited slightly enhanced working memory performance in adulthood, and better performance positively correlated with total THC self-administered during adolescence. Adolescent THC-exposed rats exhibited reductions in CB1, GABA, and glutamate receptor protein, primarily in the prefrontal cortex, dorsal hippocampus, and ventral tegmental area. CONCLUSIONS These results suggest that THC exposure at self-administered doses can produce moderate behavioral and molecular alterations, including sex-dependent effects on working memory performance in adulthood.
Collapse
Affiliation(s)
- Sierra J Stringfield
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Suite 223, Pittsburgh, PA, 15219, USA
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA
| | - Mary M Torregrossa
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Suite 223, Pittsburgh, PA, 15219, USA.
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
43
|
Raghu G, Berk M, Campochiaro PA, Jaeschke H, Marenzi G, Richeldi L, Wen FQ, Nicoletti F, Calverley PMA. The Multifaceted Therapeutic Role of N-Acetylcysteine (NAC) in Disorders Characterized by Oxidative Stress. Curr Neuropharmacol 2021; 19:1202-1224. [PMID: 33380301 PMCID: PMC8719286 DOI: 10.2174/1570159x19666201230144109] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/27/2020] [Accepted: 12/13/2020] [Indexed: 02/08/2023] Open
Abstract
Oxidative stress, which results in the damage of diverse biological molecules, is a ubiquitous cellular process implicated in the etiology of many illnesses. The sulfhydryl-containing tripeptide glutathione (GSH), which is synthesized and maintained at high concentrations in all cells, is one of the mechanisms by which cells protect themselves from oxidative stress. N-acetylcysteine (NAC), a synthetic derivative of the endogenous amino acid L-cysteine and a precursor of GSH, has been used for several decades as a mucolytic and as an antidote to acetaminophen (paracetamol) poisoning. As a mucolytic, NAC breaks the disulfide bonds of heavily cross-linked mucins, thereby reducing mucus viscosity. In vitro, NAC has antifibrotic effects on lung fibroblasts. As an antidote to acetaminophen poisoning, NAC restores the hepatic GSH pool depleted in the drug detoxification process. More recently, improved knowledge of the mechanisms by which NAC acts has expanded its clinical applications. In particular, the discovery that NAC can modulate the homeostasis of glutamate has prompted studies of NAC in neuropsychiatric diseases characterized by impaired glutamate homeostasis. This narrative review provides an overview of the most relevant and recent evidence on the clinical application of NAC, with a focus on respiratory diseases, acetaminophen poisoning, disorders of the central nervous system (chronic neuropathic pain, depression, schizophrenia, bipolar disorder, and addiction), cardiovascular disease, contrast-induced nephropathy, and ophthalmology (retinitis pigmentosa).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Peter M. A. Calverley
- Address correspondence to this author at Clinical Science Centre, University Hospital Aintree, Longmoor Lane, Liverpool UK L9 7AL; Tel: +44 151 529 5886, Fax: +44 151 529 5888; E-mail:
| |
Collapse
|
44
|
Dukes AJ, Fowler JP, Lallai V, Pushkin AN, Fowler CD. Adolescent Cannabinoid and Nicotine Exposure Differentially Alters Adult Nicotine Self-Administration in Males and Females. Nicotine Tob Res 2020; 22:1364-1373. [PMID: 32396625 DOI: 10.1093/ntr/ntaa084] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 05/06/2020] [Indexed: 12/28/2022]
Abstract
INTRODUCTION During adolescence, exposure to nicotine or cannabis independently induces effects on neuromaturation and later cognitive function. However, the potential effect of both drugs under co-use conditions has become of increasing concern given the prevalence of e-cigarettes, legalization of cannabis, and availability of synthetic "spice" cannabinoid agonists. AIMS AND METHODS The current studies investigated the effects of exposure to a cannabinoid receptor agonist (WIN55,212-2) and/or nicotine over a discrete time period in mid-adolescence on later intravenous nicotine self-administration in adult male and female mice. We further examined whether cannabinoid agonist administration in adulthood would alter nicotine reinforcement, with either acute or chronic pairing across 7 days. RESULTS We found that adult males exhibited increased nicotine self-administration at a lower, rewarding nicotine dose following adolescent cannabinoid exposure, either alone or with nicotine coadministration. In contrast, adult females demonstrated an opposing effect in which adolescent cannabinoid and nicotine coexposure resulted in decreased nicotine intake compared with the nicotine only and control groups. Furthermore, after maintaining nicotine self-administration across sessions, pretreatment with a low dose of the cannabinoid agonist decreased nicotine intake in both male and female control mice, and this lowering effect was evidenced after both acute and chronic treatment. However, the cannabinoid agonist was ineffective in altering nicotine intake in mice previously exposed to nicotine, cannabinoid agonist, or both during adolescence. CONCLUSIONS These data provide evidence that adolescent drug exposure can alter later nicotine reinforcement in a sex-specific manner and can further modulate the effectiveness of interventions in reducing nicotine intake during adulthood. IMPLICATIONS These studies demonstrate a significant impact of nicotine, cannabinoids, or coexposure on developmental processes during adolescence. Differential effects were observed within each sex, with opposing results found for cannabinoid exposure on nicotine intake in males and females. Intriguingly, we also evidenced resistance to the lowering effects of a cannabinoid agonist on nicotine intake in adulthood based on adolescent drug exposure. Thus, these findings have important implications for our understanding of the impact of nicotine and cannabinoids (eg, Δ9-tetrahydrocannabinol (THC) and synthetic "spice" cannabinoids) during development, with further implications for the effectiveness of therapeutic interventions based on prior drug exposure in youth.
Collapse
Affiliation(s)
- Angeline J Dukes
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - James P Fowler
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - Valeria Lallai
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - Anna N Pushkin
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - Christie D Fowler
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| |
Collapse
|
45
|
Ucha M, Roura-Martínez D, Ambrosio E, Higuera-Matas A. The role of the mTOR pathway in models of drug-induced reward and the behavioural constituents of addiction. J Psychopharmacol 2020; 34:1176-1199. [PMID: 32854585 DOI: 10.1177/0269881120944159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Exposure to drugs of abuse induces neuroadaptations in critical nodes of the so-called reward systems that are thought to mediate the transition from controlled drug use to the compulsive drug-seeking that characterizes addictive disorders. These neural adaptations are likely to require protein synthesis, which is regulated, among others, by the mechanistic target of the rapamycin kinase (mTOR) signalling cascade. METHODS We have performed a narrative review of the literature available in PubMed about the involvement of the mTOR pathway in drug-reward and addiction-related phenomena. AIMS The aim of this study was to review the underlying architecture of this complex intracellular network and to discuss the alterations of its components that are evident after exposure to drugs of abuse. The aim was also to delineate the effects that manipulations of the mTOR network have on models of drug reward and on paradigms that recapitulate some of the psychological components of addiction. RESULTS There is evidence for the involvement of the mTOR pathway in the acute and rewarding effects of drugs of abuse, especially psychostimulants. However, the data regarding opiates are scarce. There is a need to use sophisticated animal models of addiction to ascertain the real role of the mTOR pathway in this pathology and not just in drug-mediated reward. The involvement of this pathway in behavioural addictions and impulsivity should also be studied in detail in the future. CONCLUSIONS Although there is a plethora of data about the modulation of mTOR by drugs of abuse, the involvement of this signalling pathway in addictive disorders requires further research.
Collapse
Affiliation(s)
- Marcos Ucha
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - David Roura-Martínez
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - Emilio Ambrosio
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - Alejandro Higuera-Matas
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| |
Collapse
|
46
|
Reece AS, Hulse GK. Cannabis and Pregnancy Don't Mix. MISSOURI MEDICINE 2020; 117:530-531. [PMID: 33311778 PMCID: PMC7721409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Affiliation(s)
- Albert Stuart Reece
- Division of Psychiatry, University of Western Australia, Crawley, Western Australia & School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia
| | - Gary Kenneth Hulse
- Division of Psychiatry, University of Western Australia, Crawley, Western Australia & School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia
| |
Collapse
|
47
|
Neuhofer D, Spencer SM, Chioma VC, Beloate LN, Schwartz D, Kalivas PW. The loss of NMDAR-dependent LTD following cannabinoid self-administration is restored by positive allosteric modulation of CB1 receptors. Addict Biol 2020; 25:e12843. [PMID: 31733097 PMCID: PMC7962172 DOI: 10.1111/adb.12843] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 02/04/2023]
Abstract
Glutamatergic plasticity in the nucleus accumbens core (NAcore) is a key neuronal process in appetitive learning and contributes to pathologies such as drug addiction. Understanding how this plasticity factors into cannabis addiction and relapse has been hampered by the lack of a rodent model of cannabis self-administration. We used intravenous self-administration of two constituents of cannabis, Δ9 -tetrahydrocannabinol (THC) and cannabidiol (CBD) to examine how contingent cannabis use and cue-induced cannabinoid-seeking alters glutamatergic neurotransmission and synaptic plasticity in NAcore. NMDA receptor (NMDAR)-dependent long-term depression (LTD) in the NAcore was lost after cannabinoid, but not sucrose self-administration. Surprisingly, when rats underwent cue-induced cannabinoid seeking, LTD was restored. Loss of LTD was accompanied by desensitization of cannabinoid receptor 1 (CB1R). CB1R are positioned to regulate synaptic plasticity by being expressed on glutamatergic terminals and negatively regulating presynaptic excitability and glutamate release. Supporting this possibility, LTD was restored by promoting CB1R signaling with the CB1 positive allosteric modulator GAT211. These data implicate NAcore CB1R as critical regulators of metaplasticity induced by cannabis self-administration and the cues predicting cannabis availability.
Collapse
Affiliation(s)
- Daniela Neuhofer
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
| | - Sade M. Spencer
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
- Department of Pharmacology, Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Vivian C. Chioma
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
| | - Lauren N. Beloate
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
| | - Danielle Schwartz
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
| | - Peter W. Kalivas
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
48
|
Maternal Exposure to the Cannabinoid Agonist WIN 55,12,2 during Lactation Induces Lasting Behavioral and Synaptic Alterations in the Rat Adult Offspring of Both Sexes. eNeuro 2020; 7:ENEURO.0144-20.2020. [PMID: 32868310 PMCID: PMC7540927 DOI: 10.1523/eneuro.0144-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/09/2020] [Accepted: 07/27/2020] [Indexed: 11/30/2022] Open
Abstract
Consumption of cannabis during pregnancy and the lactation period is a rising public health concern (Scheyer et al., 2019). Exposure to synthetic or plant-derived cannabinoids via lactation disrupts the development of GABAergic neurons in the prefrontal cortex (PFC) and alters early-life behaviors (Scheyer et al., 2020b). Recently, additional data revealed that Δ9-tetrahydrocannabinol (THC) perinatal exposure via lactation causes lasting behavioral and neuronal consequences (Scheyer et al., 2020a). Here, the long-term effects in adult offspring of maternal exposure to the synthetic cannabinoid agonist WIN 55,12,2 are reported. The data demonstrate that rats exposed during lactation to WIN display social and motivational deficits at adulthood. These behavioral changes were paralleled by a specific loss of endocannabinoid-mediated long-term depression (eCB-LTD) in the PFC and nucleus accumbens (NAc), while other forms of synaptic plasticity remained intact. Thus, similarly to THC, perinatal WIN exposure via lactation induces behavioral and synaptic abnormalities lasting into adulthood.
Collapse
|
49
|
Avchalumov Y, Trenet W, Piña-Crespo J, Mandyam C. SCH23390 Reduces Methamphetamine Self-Administration and Prevents Methamphetamine-Induced Striatal LTD. Int J Mol Sci 2020; 21:E6491. [PMID: 32899459 PMCID: PMC7554976 DOI: 10.3390/ijms21186491] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022] Open
Abstract
Extended-access methamphetamine self-administration results in unregulated intake of the drug; however, the role of dorsal striatal dopamine D1-like receptors (D1Rs) in the reinforcing properties of methamphetamine under extended-access conditions is unclear. Acute (ex vivo) and chronic (in vivo) methamphetamine exposure induces neuroplastic changes in the dorsal striatum, a critical region implicated in instrumental learning. For example, methamphetamine exposure alters high-frequency stimulation (HFS)-induced long-term depression in the dorsal striatum; however, the effect of methamphetamine on HFS-induced long-term potentiation (LTP) in the dorsal striatum is unknown. In the current study, dorsal striatal infusion of SCH23390, a D1R antagonist, prior to extended-access methamphetamine self-administration reduced methamphetamine addiction-like behavior. Reduced behavior was associated with reduced expression of PSD-95 in the dorsal striatum. Electrophysiological findings demonstrate that superfusion of methamphetamine reduced basal synaptic transmission and HFS-induced LTP in dorsal striatal slices, and SCH23390 prevented this effect. These results suggest that alterations in synaptic transmission and synaptic plasticity induced by acute methamphetamine via D1Rs could assist with methamphetamine-induced modification of corticostriatal circuits underlying the learning of goal-directed instrumental actions and formation of habits, mediating escalation of methamphetamine self-administration and methamphetamine addiction-like behavior.
Collapse
Affiliation(s)
- Yosef Avchalumov
- VA San Diego Healthcare System, San Diego, CA 92161, USA; (Y.A.); (W.T.)
| | - Wulfran Trenet
- VA San Diego Healthcare System, San Diego, CA 92161, USA; (Y.A.); (W.T.)
| | - Juan Piña-Crespo
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA;
| | - Chitra Mandyam
- VA San Diego Healthcare System, San Diego, CA 92161, USA; (Y.A.); (W.T.)
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| |
Collapse
|
50
|
Oxidative Stress and Neuroinflammation as a Pivot in Drug Abuse. A Focus on the Therapeutic Potential of Antioxidant and Anti-Inflammatory Agents and Biomolecules. Antioxidants (Basel) 2020; 9:antiox9090830. [PMID: 32899889 PMCID: PMC7555323 DOI: 10.3390/antiox9090830] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
Drug abuse is a major global health and economic problem. However, there are no pharmacological treatments to effectively reduce the compulsive use of most drugs of abuse. Despite exerting different mechanisms of action, all drugs of abuse promote the activation of the brain reward system, with lasting neurobiological consequences that potentiate subsequent consumption. Recent evidence shows that the brain displays marked oxidative stress and neuroinflammation following chronic drug consumption. Brain oxidative stress and neuroinflammation disrupt glutamate homeostasis by impairing synaptic and extra-synaptic glutamate transport, reducing GLT-1, and system Xc− activities respectively, which increases glutamatergic neurotransmission. This effect consolidates the relapse-promoting effect of drug-related cues, thus sustaining drug craving and subsequent drug consumption. Recently, promising results as experimental treatments to reduce drug consumption and relapse have been shown by (i) antioxidant and anti-inflammatory synthetic molecules whose effects reach the brain; (ii) natural biomolecules secreted by mesenchymal stem cells that excel in antioxidant and anti-inflammatory properties, delivered via non-invasive intranasal administration to animal models of drug abuse and (iii) potent anti-inflammatory microRNAs and anti-miRNAs which target the microglia and reduce neuroinflammation and drug craving. In this review, we address the neurobiological consequences of brain oxidative stress and neuroinflammation that follow the chronic consumption of most drugs of abuse, and the current and potential therapeutic effects of antioxidants and anti-inflammatory agents and biomolecules to reduce these drug-induced alterations and to prevent relapse.
Collapse
|