1
|
Larcombe AN, Chivers EK, Landwehr KR, Berry LJ, de Jong E, Huxley RR, Musk A, Franklin PJ, Mullins BJ. Partial amelioration of a chronic cigarette-smoke-induced phenotype in mice by switching to electronic cigarettes. Arch Toxicol 2025:10.1007/s00204-025-04055-7. [PMID: 40249508 DOI: 10.1007/s00204-025-04055-7] [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: 01/22/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025]
Abstract
Electronic cigarettes ("e-cigarettes") are often marketed as smoking cessation tools and are used by smokers to reduce/quit cigarette smoking. The objective of this study was to assess the health effects of switching to e-cigarettes after long-term smoking in a mouse model and compare these effects with continued smoking, or quitting entirely. Adult BALB/c mice were whole-body exposed to mainstream cigarette smoke (2 h/day, 5 days/week) for 12 weeks prior to switching to flavoured e-cigarette aerosol (50:50 propylene glycol and glycerine) containing 18 mg/mL nicotine (2 h/day and 5 days/week), continuing cigarette smoking (2 h/day and 5 days/week), or quitting entirely for an additional 2 weeks. We then assessed a range of respiratory health outcomes including lung function and structure, pulmonary inflammation and changes in gene expression in the lung. Switching to e-cigarettes led to improvements in some aspects of respiratory health in mice compared with continued smoking, such as reduced neutrophilic inflammation in the lung. However, total cellular lung inflammation was still elevated and lung function was still impaired, in terms of airway responsiveness to methacholine, for e-cigarette use compared with quitting. Larger effects were typically seen in female mice compared to male. This study shows that switching to e-cigarettes after long-term cigarette smoking leads to improvements in some aspects of respiratory health, such as neutrophilic inflammation and the volume dependence of lung function compared with continued smoking. However, switching to e-cigarettes was not as effective as quitting smoking entirely.
Collapse
Affiliation(s)
- Alexander N Larcombe
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia.
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA, Australia.
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| | - Emily K Chivers
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
| | - Katherine R Landwehr
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA, Australia
| | - Luke J Berry
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
| | - Emma de Jong
- Centre for Health Research, The Kids Research Institute Australia, The University of Western Australia, Perth, WA, Australia
| | - Rachel R Huxley
- The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
- Faculty of Health, Deakin University, Geelong, VIC, Australia
| | - Arthur Musk
- School of Population and Global Health, University of Western Australia, Perth, WA, Australia
| | - Peter J Franklin
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
- School of Population and Global Health, University of Western Australia, Perth, WA, Australia
| | - Benjamin J Mullins
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA, Australia
| |
Collapse
|
2
|
Mabou Tagne A, Fotio Y, Gupta K, Piomelli D. Δ 9-Tetrahydrocannabinol Alleviates Hyperalgesia in a Humanized Mouse Model of Sickle Cell Disease. J Pharmacol Exp Ther 2024; 391:174-181. [PMID: 38955494 DOI: 10.1124/jpet.124.002285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
People with sickle cell disease (SCD) often experience chronic pain as well as unpredictable episodes of acute pain, which significantly affects their quality of life and life expectancy. Current treatment strategies for SCD-associated pain primarily rely on opioid analgesics, which have limited efficacy and cause serious adverse effects. Cannabis has emerged as a potential alternative, yet its efficacy remains uncertain. In this study, we investigated the antinociceptive effects of Δ9-tetrahydrocannabinol (THC), cannabis' intoxicating constituent, in male HbSS mice, which express >99% human sickle hemoglobin, and male HbAA mice, which express normal human hemoglobin A, as a control. Acute THC administration (0.1-3 mg/kg-1, i.p.) dose-dependently reduced mechanical and cold hypersensitivity in human sickle hemoglobin (HbSS) but not human normal hemoglobin A (HbAA) mice. In the tail-flick assay, THC (1 and 3 mg/kg-1, i.p.) produced substantial antinociceptive effects in HbSS mice. By contrast, THC (1 mg/kg-1, i.p.) did not alter anxiety-like behavior (elevated plus maze) or long-term memory (24-hour novel object recognition). Subchronic THC treatment (1 and 3 mg/kg-1, i.p.) provided sustained relief of mechanical hypersensitivity but led to tolerance in cold hypersensitivity in HbSS mice. Together, the findings identify THC as a possible therapeutic option for the management of chronic pain in SCD. Further research is warranted to elucidate its mechanism of action and possible interaction with other cannabis constituents. SIGNIFICANCE STATEMENT: The study explores Δ9-tetrahydrocannabinol (THC)'s efficacy in alleviating pain in sickle cell disease (SCD) using a humanized mouse model. Findings indicate that acute THC administration reduces mechanical and cold hypersensitivity in SCD mice without impacting emotional and cognitive dysfunction. Subchronic THC treatment offers sustained relief of mechanical hypersensitivity but leads to cold hypersensitivity tolerance. These results offer insights into THC's potential as an alternative pain management option in SCD, highlighting both its benefits and limitations.
Collapse
Affiliation(s)
- Alex Mabou Tagne
- Departments of Anatomy and Neurobiology (A.M.T., Y.F., D.P.), Biological Chemistry (D.P.), and Pharmaceutical Sciences (D.P.), and Hematology/Oncology, Department of Medicine (K.G.), University of California Irvine, Irvine, California
| | - Yannick Fotio
- Departments of Anatomy and Neurobiology (A.M.T., Y.F., D.P.), Biological Chemistry (D.P.), and Pharmaceutical Sciences (D.P.), and Hematology/Oncology, Department of Medicine (K.G.), University of California Irvine, Irvine, California
| | - Kalpna Gupta
- Departments of Anatomy and Neurobiology (A.M.T., Y.F., D.P.), Biological Chemistry (D.P.), and Pharmaceutical Sciences (D.P.), and Hematology/Oncology, Department of Medicine (K.G.), University of California Irvine, Irvine, California
| | - Daniele Piomelli
- Departments of Anatomy and Neurobiology (A.M.T., Y.F., D.P.), Biological Chemistry (D.P.), and Pharmaceutical Sciences (D.P.), and Hematology/Oncology, Department of Medicine (K.G.), University of California Irvine, Irvine, California
| |
Collapse
|
3
|
Lallai V, Lam TT, Garcia-Milian R, Chen YC, Fowler JP, Manca L, Piomelli D, Williams K, Nairn AC, Fowler CD. Proteomic Profile of Circulating Extracellular Vesicles in the Brain after Δ9-Tetrahydrocannabinol Inhalation. Biomolecules 2024; 14:1143. [PMID: 39334909 PMCID: PMC11430348 DOI: 10.3390/biom14091143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Given the increasing use of cannabis in the US, there is an urgent need to better understand the drug's effects on central signaling mechanisms. Extracellular vesicles (EVs) have been identified as intercellular signaling mediators that contain a variety of cargo, including proteins. Here, we examined whether the main psychoactive component in cannabis, Δ9-tetrahydrocannabinol (THC), alters EV protein signaling dynamics in the brain. We first conducted in vitro studies, which found that THC activates signaling in choroid plexus epithelial cells, resulting in transcriptional upregulation of the cannabinoid 1 receptor and immediate early gene c-fos, in addition to the release of EVs containing RNA cargo. Next, male and female rats were examined for the effects of either acute or chronic exposure to aerosolized ('vaped') THC on circulating brain EVs. Cerebrospinal fluid was extracted from the brain, and EVs were isolated and processed with label-free quantitative proteomic analyses via high-resolution tandem mass spectrometry. Interestingly, circulating EV-localized proteins were differentially expressed based on acute or chronic THC exposure in a sex-specific manner. Taken together, these findings reveal that THC acts in the brain to modulate circulating EV signaling, thereby providing a novel understanding of how exogenous factors can regulate intercellular communication in the brain.
Collapse
Affiliation(s)
- Valeria Lallai
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
| | - TuKiet T. Lam
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT 06511, USA
| | - Rolando Garcia-Milian
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Bioinformatics Support Hub, Harvey Cushing/John Whitney Medical Library, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yen-Chu Chen
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
| | - James P. Fowler
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
| | - Letizia Manca
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
| | - Daniele Piomelli
- Department and Anatomy and Neurobiology, University of California, Irvine, CA 92697, USA;
| | - Kenneth Williams
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Angus C. Nairn
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Department of Psychiatry, Yale University, New Haven, CT 06511, USA
| | - Christie D. Fowler
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
| |
Collapse
|
4
|
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
|
5
|
Sicher AR, Liss A, Vozella V, Marsland P, Seemiller LR, Springer M, Starnes WD, Griffith KR, Smith GC, Astefanous A, Deak T, Roberto M, Varodayan FP, Crowley NA. Voluntary adolescent alcohol exposure does not robustly increase adulthood consumption of alcohol in multiple mouse and rat models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591674. [PMID: 38746266 PMCID: PMC11092607 DOI: 10.1101/2024.04.30.591674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Adolescence is a period of increased risk taking, including increased alcohol and drug use. Multiple clinical studies report a positive relationship between adolescent alcohol consumption and risk of developing an alcohol use disorder (AUD) in adulthood. However, few preclinical studies have attempted to tease apart the biological contributions of adolescent alcohol exposure, independent of other social, environmental, and stress factors, and studies that have been conducted show mixed results. Here we use several adolescent voluntary consumption of alcohol models, conducted across four labs in three institutes and with two rodent species, to investigate the ramifications of adolescent alcohol consumption on adulthood alcohol consumption in controlled, pre-clinical environments. We consistently demonstrate a lack of robust increases in adulthood alcohol consumption. This work highlights that risks seen in both human datasets and other murine drinking models may be due to unique social and environmental factors - some of which may be unique to humans.
Collapse
Affiliation(s)
- Avery R. Sicher
- Neuroscience Graduate Program, The Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Andrea Liss
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University - SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Valentina Vozella
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Paige Marsland
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University - SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Laurel R. Seemiller
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Matthew Springer
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - William D. Starnes
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Keith R. Griffith
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Grace C. Smith
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Amy Astefanous
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University - SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University - SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Florence P. Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University - SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Nicole A. Crowley
- Neuroscience Graduate Program, The Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
6
|
Xu Y, Li X, Xu P, Yan F, Wang D. Comparative pharmacokinetic and intracerebral distribution of MDMB-4F-BICA in mice following inhalation ('vapor') and subcutaneous injection. J Pharm Biomed Anal 2024; 241:115988. [PMID: 38301574 DOI: 10.1016/j.jpba.2024.115988] [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: 10/25/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 02/03/2024]
Abstract
MDMB-4F-BICA, also known as 4F-MDMB-BICA, is a new psychoactive substance that emerged in 2020. It is often illegally added to electronic cigarette oil for inhalation abuse, leading to serious adverse symptoms and even death. There are significant differences in pharmacokinetics between inhalation administration and conventional drug delivery methods. Inhalation administration can pass through the blood-brain barrier to enter the brain directly. However, the specific distribution of the drug in the brain following inhalation has not been well investigated. In order to scientifically compare the absorption and distribution of MDMB-4F-BICA after two administration methods (inhalation and subcutaneous injection), this study analyzed the drug concentration in mice blood and brain by LC-MS/MS after systemic exposure inhalation in the form of electronic cigarettes. The aim was to conduct the pharmacokinetics study of MDMB-4F-BICA after inhalation('vapor') administration. Pharmacokinetics and distribution of the compound revealed that the maximum concentrations in blood of this compound were reached at 0.5 min and 15 min, respectively, and the concentration in the brain reached the maximum at the same time after two modes of administration. The drug concentration in the brain was higher than that of subcutaneous injection, and the drug remained at a low concentration in the brain for a long period (20 ng/g brain tissue) with a significant distribution in several olfactory primary cortex brain regions. Taken together, the pharmacokinetics of the synthetic cannabinoid MDMB-4F-BICA after single systemic exposure inhalation were investigated for the first time in this study. A basis for subsequent evaluation research of inhalation-related harmfulness is provided by comparing the distribution of drugs in the brain after the two administration modes.
Collapse
Affiliation(s)
- Yawen Xu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Beijing 100193, China
| | - Xiangyu Li
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100193, China; Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Beijing 100193, China
| | - Peng Xu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100193, China; Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Beijing 100193, China
| | - Fang Yan
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Beijing 100193, China.
| | - Dan Wang
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100193, China; Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Beijing 100193, China.
| |
Collapse
|
7
|
Lim J, Lee HL, Nguyen J, Shin J, Getze S, Quach C, Squire E, Jung KM, Mahler SV, Mackie K, Piomelli D, Luderer U. Adolescent exposure to low-dose Δ9-tetrahydrocannabinol depletes the ovarian reserve in female mice. Toxicol Sci 2023; 193:31-47. [PMID: 36912754 PMCID: PMC10176244 DOI: 10.1093/toxsci/kfad027] [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: 03/14/2023] Open
Abstract
Cannabis use by adolescents is widespread, but its effects on the ovaries remain largely unknown. Δ9-tetrahydrocannabinol (THC) exerts its pharmacological effects by activating, and in some conditions hijacking, cannabinoid receptors (CBRs). We hypothesized that adolescent exposure to THC affects ovarian function in adulthood. Peripubertal female C57BL/6N mice were given THC (5 mg/kg) or its vehicle, once daily by intraperitoneal injection. Some mice received THC from postnatal day (PND) 30-33 and their ovaries were harvested PND34; other mice received THC from PND30-43, and their ovaries were harvested PND70. Adolescent treatment with THC depleted ovarian primordial follicle numbers by 50% at PND70, 4 weeks after the last dose. The treatment produced primordial follicle activation, which persisted until PND70. THC administration also caused DNA damage in primary follicles and increased PUMA protein expression in oocytes of primordial and primary follicles. Both CB1R and CB2R were expressed in oocytes and theca cells of ovarian follicles. Enzymes involved in the formation (N-acylphosphatidylethanolamine phospholipase D) or deactivation (fatty acid amide hydrolase) of the endocannabinoid anandamide were expressed in granulosa cells of ovarian follicles and interstitial cells. Levels of mRNA for CBR1 were significantly increased in ovaries after adolescent THC exposure, and upregulation persisted for at least 4 weeks. Our results support that adolescent exposure to THC may cause aberrant activation of the ovarian endocannabinoid system in female mice, resulting in substantial loss of ovarian reserve in adulthood. Relevance of these findings to women who frequently used cannabis during adolescence warrants investigation.
Collapse
Affiliation(s)
- Jinhwan Lim
- Department of Environmental and Occupational Health, University of California Irvine, Irvine, California 92697, USA
- Dept. of Medicine, University of California Irvine, Irvine, California 92697, USA
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California 92697, USA
| | - Julie Nguyen
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California 92697, USA
| | - Joyce Shin
- Department of Environmental and Occupational Health, University of California Irvine, Irvine, California 92697, USA
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California 92697, USA
| | - Samantha Getze
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California 92697, USA
| | - Caitlin Quach
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California 92697, USA
| | - Erica Squire
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California 92697, USA
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California 92697, USA
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California 92697, USA
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California 92697, USA
| | - Ulrike Luderer
- Department of Environmental and Occupational Health, University of California Irvine, Irvine, California 92697, USA
- Dept. of Medicine, University of California Irvine, Irvine, California 92697, USA
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California 92697, USA
| |
Collapse
|
8
|
Del Rio R, Serrano RG, Gomez E, Martinez JC, Edward MA, Santos RA, Diaz KS, Cohen-Cory S. Cell-autonomous and differential endocannabinoid signaling impacts the development of presynaptic retinal ganglion cell axon connectivity in vivo. Front Synaptic Neurosci 2023; 15:1176864. [PMID: 37252636 PMCID: PMC10213524 DOI: 10.3389/fnsyn.2023.1176864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
Cannabis exposure during gestation evokes significant molecular modifications to neurodevelopmental programs leading to neurophysiological and behavioral abnormalities in humans. The main neuronal receptor for Δ9-tetrahydrocannabinol (THC) is the type-1 cannabinoid receptor CB1R, one of the most abundant G-protein-coupled receptors in the nervous system. While THC is the major psychoactive phytocannabinoid, endocannabinoids (eCBs) are the endogenous ligands of CB1R and are known to act as retrograde messengers to modulate synaptic plasticity at different time scales in the adult brain. Accumulating evidence indicates that eCB signaling through activation of CB1R plays a central role in neural development. During development, most CB1R localized to axons of projection neurons, and in mice eCB signaling impacts axon fasciculation. Understanding of eCB-mediated structural plasticity during development, however, requires the identification of the precise spatial and temporal dynamics of CB1R-mediated modifications at the level of individual neurons in the intact brain. Here, the cell-autonomous role of CB1R and the effects of CB1R-mediated eCB signaling were investigated using targeted single-cell knockdown and pharmacologic treatments in Xenopus. We imaged axonal arbors of retinal ganglion cells (RGCs) in real time following downregulation of CB1R via morpholino (MO) knockdown. We also analyzed RGC axons with altered eCB signaling following treatment with URB597, a selective inhibitor of the enzyme that degrades Anandamide (AEA), or JZL184, an inhibitor of the enzyme that blocks 2-Arachidonoylglycerol (2-AG) hydrolysis, at two distinct stages of retinotectal development. Our results demonstrate that CB1R knockdown impacts RGC axon branching at their target and that differential 2-AG and AEA-mediated eCB signaling contributes to presynaptic structural connectivity at the time that axons terminate and when retinotectal synaptic connections are made. Altering CB1R levels through CB1R MO knockdown similarly impacted dendritic morphology of tectal neurons, thus supporting both pre- and postsynaptic cell-autonomous roles for CB1R-mediated eCB signaling.
Collapse
|
9
|
Tang TYC, Kim JS, Das A. Role of omega-3 and omega-6 endocannabinoids in cardiopulmonary pharmacology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 97:375-422. [PMID: 37236765 DOI: 10.1016/bs.apha.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Endocannabinoids are derived from dietary omega-3 and omega-6 fatty acids and play an important role in regulation of inflammation, development, neurodegenerative diseases, cancer, and cardiovascular diseases. They elicit this effect via interactions with cannabinoid receptors 1 and 2 which are also targeted by plant derived cannabinoid from cannabis. The evidence of the involvement of the endocannabinoid system in cardiopulmonary function comes from studies that show that cannabis consumption leads to cardiovascular effect such as arrythmia and is beneficial in lung cancer patients. Moreover, omega-3 and omega-6 endocannabinoids play several important roles in cardiopulmonary system such as causing airway relaxation, suppressing atherosclerosis and hypertension. These effects are mediated via the cannabinoids receptors that are abundant in the cardiopulmonary system. Overall, this chapter reviews the known role of phytocannabinoids and endocannabinoids in the cardiopulmonary context.
Collapse
Affiliation(s)
- Tiffany Y-C Tang
- School of Chemistry and Biochemistry, College of Sciences. Georgia Institute of Technology, Parker H. Petit Institute for Bioengineering and Biosciences, Atlanta, GA, United States
| | - Justin S Kim
- School of Chemistry and Biochemistry, College of Sciences. Georgia Institute of Technology, Parker H. Petit Institute for Bioengineering and Biosciences, Atlanta, GA, United States
| | - Aditi Das
- School of Chemistry and Biochemistry, College of Sciences. Georgia Institute of Technology, Parker H. Petit Institute for Bioengineering and Biosciences, Atlanta, GA, United States.
| |
Collapse
|