1
|
Arnold JC, Occelli Hanbury-Brown CV, Anderson LL, Bedoya-Pérez MA, Udoh M, Sharman LA, Raymond JS, Doohan PT, Ametovski A, McGregor IS. A sleepy cannabis constituent: cannabinol and its active metabolite influence sleep architecture in rats. Neuropsychopharmacology 2025; 50:586-595. [PMID: 39528623 PMCID: PMC11736144 DOI: 10.1038/s41386-024-02018-7] [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: 08/08/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Medicinal cannabis is being used worldwide and there is increasing use of novel cannabis products in the community. Cannabis contains the major cannabinoids, Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD), but also an array of minor cannabinoids that have undergone much less pharmacological characterization. Cannabinol (CBN) is a minor cannabinoid used in the community in "isolate' products and is claimed to have pro-sleep effects comparable to conventional sleep medications. However, no study has yet examined whether it impacts sleep architecture using objective sleep measures. The effects of CBN on sleep in rats using polysomnography were therefore examined. CBN increased total sleep time, although there was evidence of biphasic effects with initial sleep suppression before a dramatic increase in sleep. CBN increased both non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. The magnitude of the effect of CBN on NREM was comparable to the sleep aid zolpidem, although, unlike CBN, zolpidem did not influence REM sleep. Following CBN dosing, 11-hydroxy-CBN, a primary metabolite of CBN surprisingly attained equivalently high brain concentrations to CBN. 11-hydroxy-CBN was active at cannabinoid CB1 receptors with comparable potency and efficacy to Δ9-THC, however, CBN had much lower activity. We then discovered that the metabolite 11-hydroxy-CBN also influenced sleep architecture, albeit with some subtle differences from CBN itself. This study shows CBN affects sleep using objective sleep measures and suggests an active metabolite may contribute to its hypnotic action.
Collapse
Affiliation(s)
- Jonathon C Arnold
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia.
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
| | - Cassandra V Occelli Hanbury-Brown
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Lyndsey L Anderson
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Miguel A Bedoya-Pérez
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Michael Udoh
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Laura A Sharman
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Joel S Raymond
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Peter T Doohan
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Adam Ametovski
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Iain S McGregor
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
2
|
Xue J, Xu Z, Zhang J, Hou H, Ge L, Yang K. Systematic review/meta-analysis on the role of CB1R regulation in sleep-wake cycle in rats. J Evid Based Med 2024; 17:714-728. [PMID: 39325651 DOI: 10.1111/jebm.12643] [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: 05/15/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024]
Abstract
OBJECTIVE A systematic review/meta-analysis was conducted to investigate the effect of cannabinoid type-1 receptor (CB1R) regulation on the sleep-wake cycle of rats and to provide new ideas and evidence-based basis for clinical research on the treatment of sleep disorders. METHODS We searched Cochrane Library, PubMed, Web of Science, Embase, Chinese Biomedicine Literature Database (CBM), China National Knowledge Infrastructure, WanFang, and VIP databases for relevant papers, about the effects of CB1R agonists/antagonists on sleep-wake cycle in rats, from inception to November 2023. Two reviewers performed study screening, data extraction, and risk of bias assessment using the SYRCLE's risk of bias tool. Meta-analysis was performed using RevMan 5.3 software. Heterogeneity test was performed on the included studies (Test standard α = 0.1). I2 value was used to evaluate the heterogeneity. Forest plot was drawn, and p ≤ 0.05 indicates statistically significant difference. RESULTS A total of 16 trials involving 484 experimental rats were included. The methodological quality evaluation results showed that the overall quality of the included studies was low. The results of the meta-analysis showed that single administration of CB1R agonists could shorten the wakefulness (W) time in the first 6 h (h) (standardized mean difference (SMD) = -2.52, 95% confidence interval (CI) (-3.83, -1.22), p = 0.0002) and 24 h (SMD = -0.84, 95% CI (-1.31, -0.36), p = 0.0005) after administration, prolong nonrapid eye movement sleep (NREM) time (SMD = 1.75, 95% CI (0.54, 2.95), p = 0.005) and rapid eye movement sleep (REM) time (SMD = 1.76, 95% CI (0.26, 3.26), p = 0.02), and increase REM frequency after administration (SMD = 1.67, 95% CI (0.98, 2.35), p < 0.00001), these results were all statistically different. There were no significant differences in sleep latency and average duration of REM. Single administration of CB1R antagonists prolonged the first 6 h W time after administration (SMD = 1.36, 95%CI (0.29, 2.43), p = 0.01), shortened the first 6 h NREM time (SMD = -1.73, 95% CI (-2.88, -0.57), p = 0.003) and REM time (SMD = -2.07, 95% CI (-3.17, -0.96), p = 0.0003) after administration, and increased the frequency of W after administration (SMD = 3.57, 95% CI (1.42, 5.72), p = 0.001). There was no statistical difference in the average duration of W. REM time and REM frequency increased after continuous CB1R agonist withdrawal. CONCLUSIONS According to the existing evidence, CB1R played a pivotal role in regulating the sleep-wake cycle in rats. CB1R agonists tended to reduce W time, increase NREM and REM sleep times, boost REM frequency, and promote sleep. Conversely, CB1R antagonists could increase the duration and frequency of W, shorten NREM and REM sleep times, and promote W.
Collapse
Affiliation(s)
- Jianjun Xue
- Center for Evidence-Based Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
- Gansu Province Clinical Research Center of Integrative Anaesthesiology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
- Anesthesiology and Pain Medicine Center, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Ziqing Xu
- Gansu Province Clinical Research Center of Integrative Anaesthesiology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
- Anesthesiology and Pain Medicine Center, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Jie Zhang
- Gansu Province Clinical Research Center of Integrative Anaesthesiology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
- Anesthesiology and Pain Medicine Center, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Huaijing Hou
- Gansu Province Clinical Research Center of Integrative Anaesthesiology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
- Anesthesiology and Pain Medicine Center, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Long Ge
- Center for Evidence-Based Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Kehu Yang
- Center for Evidence-Based Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
- Center for Evidence-Based Social Science/Center for Health Technology Assessment, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, Gansu, China
- Chinese GRADE Center, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
3
|
Gamble MC, Miracle S, Williams BR, Logan RW. Endocannabinoid agonist 2-arachidonoylglycerol differentially alters diurnal activity and sleep during fentanyl withdrawal in male and female mice. Pharmacol Biochem Behav 2024; 240:173791. [PMID: 38761993 PMCID: PMC11166043 DOI: 10.1016/j.pbb.2024.173791] [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: 12/21/2023] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Fentanyl has become the leading driver of opioid overdoses in the United States. Cessation of opioid use represents a challenge as the experience of withdrawal drives subsequent relapse. One of the most prominent withdrawal symptoms that can contribute to opioid craving and vulnerability to relapse is sleep disruption. The endocannabinoid agonist, 2-Arachidonoylglycerol (2-AG), may promote sleep and reduce withdrawal severity; however, the effects of 2-AG on sleep disruption during opioid withdrawal have yet to be assessed. Here, we investigated the effects of 2-AG administration on sleep-wake behavior and diurnal activity in mice during withdrawal from fentanyl. Sleep-wake activity measured via actigraphy was continuously recorded before and after chronic fentanyl administration in both male and female C57BL/6J mice. Immediately following cessation of fentanyl administration, 2-AG was administered intraperitoneally to investigate the impact of endocannabinoid agonism on opioid-induced sleep disruption. We found that female mice maintained higher activity levels in response to chronic fentanyl than male mice. Furthermore, fentanyl administration increased wake and decreased sleep during the light period and inversely increased sleep and decreased wake in the dark period in both sexes. 2-AG treatment increased arousal and decreased sleep in both sexes during first 24-h of withdrawal. On withdrawal day 2, only females showed increased wakefulness with no changes in males, but by withdrawal day 3 male mice displayed decreased rapid-eye movement sleep during the dark period with no changes in female mice. Overall, repeated administration of fentanyl altered sleep and diurnal activity and administration of the endocannabinoid agonist, 2-AG, had sex-specific effects on fentanyl-induced sleep and diurnal changes.
Collapse
Affiliation(s)
- Mackenzie C Gamble
- Molecular and Translational Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Sophia Miracle
- Graduate Program in Neuroscience, Boston University, Boston, MA, USA
| | - Benjamin R Williams
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ryan W Logan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
4
|
Belali R, Mard SA, Khoshnam SE, Bavarsad K, Sarkaki A, Farbood Y. Anandamide Attenuates Neurobehavioral Deficits and EEG Irregularities in the Chronic Sleep Deprivation Rats: The Role of Oxidative Stress and Neuroinflammation. Neurochem Res 2024; 49:1541-1555. [PMID: 37966567 DOI: 10.1007/s11064-023-04054-z] [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] [Received: 04/28/2023] [Revised: 10/10/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023]
Abstract
Sleep deprivation increases stress, anxiety, and depression by altering the endocannabinoid system's function. In the present study, we aimed to investigate the anti-anxiety and anti-depressant effects of the endocannabinoid anandamide (AEA) in the chronic sleep deprivation (SD) model in rats. Adult male Wistar rats (200-250 g) were randomly divided into three groups: control + vehicle (Control), chronic sleep deprivation + vehicle (SD), and chronic sleep deprivation + 20 mg/kg AEA (SD + A). The rats were kept in a sleep deprivation device for 18 h (7 to 1 a.m.) daily for 21 days. Open-field (OFT), elevated plus maze, and forced swimming tests (FST) were used to assess anxiety and depression-like behavior. As well as the cortical EEG, CB1R mRNA expression, TNF-α, IL-6, IL-4 levels, and antioxidant activity in the brain were examined following SD induction. AEA administration significantly increased the time spent (p < 0.01), the distance traveled in the central zone (p < 0.001), and the number of climbing (p < 0.05) in the OFT; it also increased the duration and number of entries into the open arms (p < 0.01 and p < 0.05 respectively), and did not reduce immobility time in the FST (p > 0.05), AEA increased CB1R mRNA expression in the anterior and medial parts of the brain (p < 0.01), and IL-4 levels (p < 0.05). AEA also reduced IL-6 and TNF-α (p < 0.05) and modulated cortical EEG. AEA induced anxiolytic-like effects but not anti-depressant effects in the SD model in rats by modulating CB1R mRNA expression, cortical EEG, and inflammatory response.
Collapse
Affiliation(s)
- Rafie Belali
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Ali Mard
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kowsar Bavarsad
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Sarkaki
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Yaghoob Farbood
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
5
|
Martin SC, Joyce KK, Harper KM, Harp SJ, Cohen TJ, Moy SS, Diering GH. Evaluating Fatty Acid Amide Hydrolase as a Suitable Target for Sleep Promotion in a Transgenic TauP301S Mouse Model of Neurodegeneration. Pharmaceuticals (Basel) 2024; 17:319. [PMID: 38543105 PMCID: PMC10975243 DOI: 10.3390/ph17030319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 04/01/2024] Open
Abstract
Sleep disruption is an expected component of aging and neurodegenerative conditions, including Alzheimer's disease (AD). Sleep disruption has been demonstrated as a driver of AD pathology and cognitive decline. Therefore, treatments designed to maintain sleep may be effective in slowing or halting AD progression. However, commonly used sleep aid medications are associated with an increased risk of AD, highlighting the need for sleep aids with novel mechanisms of action. The endocannabinoid system holds promise as a potentially effective and novel sleep-enhancing target. By using pharmacology and genetic knockout strategies, we evaluated fatty acid amide hydrolase (FAAH) as a therapeutic target to improve sleep and halt disease progression in a transgenic Tau P301S (PS19) model of Tauopathy and AD. We have recently shown that PS19 mice exhibit sleep disruption in the form of dark phase hyperarousal as an early symptom that precedes robust Tau pathology and cognitive decline. Acute FAAH inhibition with PF3845 resulted in immediate improvements in sleep behaviors in male and female PS19 mice, supporting FAAH as a potentially suitable sleep-promoting target. Moreover, sustained drug dosing for 5-10 days resulted in maintained improvements in sleep. To evaluate the effect of chronic FAAH inhibition as a possible therapeutic strategy, we generated FAAH-/- PS19 mice models. Counter to our expectations, FAAH knockout did not protect PS19 mice from progressive sleep loss, neuroinflammation, or cognitive decline. Our results provide support for FAAH as a novel target for sleep-promoting therapies but further indicate that the complete loss of FAAH activity may be detrimental.
Collapse
Affiliation(s)
- Shenée C. Martin
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathryn K. Joyce
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathryn M. Harper
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Samuel J. Harp
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Todd J. Cohen
- Department of Neurology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sheryl S. Moy
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Institute for Developmental Disabilities, Carrboro, NC 27510, USA
| | - Graham H. Diering
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Institute for Developmental Disabilities, Carrboro, NC 27510, USA
| |
Collapse
|
6
|
Veitch MR, Jairam S, Gurges P, Cohen E, Kendzerska T, Murray BJ, Boulos MI. Cannabinoid Use and Obstructive Sleep Apnea: A Retrospective Cohort Study. Can J Neurol Sci 2024:1-8. [PMID: 38383993 DOI: 10.1017/cjn.2024.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a sleep disorder with no widely accepted pharmacological therapy. Cannabinoids have been suggested to reduce OSA severity in small human studies. The purpose of this retrospective cohort study was to explore the association of self-reported cannabis use on OSA severity and sleep parameters in a large cohort of adults undergoing in-laboratory polysomnography. METHODS Sleep and medication data were collected for all consecutive adults who completed diagnostic polysomnography at Sunnybrook Health Sciences Centre from 2010 to 2022. Multivariable linear regression models were employed that adjusted for age, sex, and BMI (minimally adjusted model), as well as medication and comorbidity data (maximally adjusted model). An exploratory subgroup analysis was additionally run in patients with moderate to severe OSA. RESULTS Of 6,958 individuals (mean age 54.7 ± 16.3, BMI 29.1 ± 6.8, 51.0% female), 71 reported cannabis use. In our minimally adjusted models, cannabis use predicted a reduced respiratory disturbance index (RDI) (β: -4.8 [95% CI: -9.4, -0.2]; p = 0.042); this association became nonsignificant in the fully adjusted models. In an exploratory analysis of patients with moderate to severe OSA (n = 613), cannabis use (n = 7) predicted increased stage N3 sleep (β: 33.5 [95% CI: 15.6, 51.4]; p < 0.001) and decreased REM sleep (β: 16.0 [95% CI: 0.3, 31.7]; p = 0.046). CONCLUSION Self-reported cannabis use was not associated with OSA severity after adjusting for confounders. In an exploratory subgroup analysis of patients with moderate to severe OSA, cannabis use impacted sleep architecture. Future studies should further explore these findings.
Collapse
Affiliation(s)
- Matthew R Veitch
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sean Jairam
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Patrick Gurges
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Elliott Cohen
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Sleep Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | - Brian J Murray
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Sleep Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Mark I Boulos
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Sleep Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| |
Collapse
|
7
|
Rice LJ, Cannon L, Dadlani N, Cheung MMY, Einfeld SL, Efron D, Dossetor DR, Elliott EJ. Efficacy of cannabinoids in neurodevelopmental and neuropsychiatric disorders among children and adolescents: a systematic review. Eur Child Adolesc Psychiatry 2024; 33:505-526. [PMID: 36864363 PMCID: PMC10869397 DOI: 10.1007/s00787-023-02169-w] [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: 09/27/2022] [Accepted: 02/14/2023] [Indexed: 03/04/2023]
Abstract
A better understanding of the endocannabinoid system and a relaxation in regulatory control of cannabis globally has increased interest in the medicinal use of cannabinoid-based products (CBP). We provide a systematic review of the rationale and current clinical trial evidence for CBP in the treatment of neuropsychiatric and neurodevelopmental disorders in children and adolescents. A systematic search of MEDLINE, Embase, PsycINFO, and the Cochrane Central Register of Trials was performed to identify articles published after 1980 about CBP for medical purposes in individuals aged 18 years or younger with selected neuropsychiatric or neurodevelopmental conditions. Risk of bias and quality of evidence was assessed for each article. Of 4466 articles screened, 18 were eligible for inclusion, addressing eight conditions (anxiety disorders (n = 1); autism spectrum disorder (n = 5); foetal alcohol spectrum disorder (n = 1); fragile X syndrome (n = 2); intellectual disability (n = 1); mood disorders (n = 2); post-traumatic stress disorder (n = 3); and Tourette syndrome (n = 3)). Only one randomised controlled trial (RCT) was identified. The remaining seventeen articles included one open-label trial, three uncontrolled before-and-after trials, two case series and 11 case reports, thus the risk of bias was high. Despite growing community and scientific interest, our systematic review identified limited and generally poor-quality evidence for the efficacy of CBP in neuropsychiatric and neurodevelopmental disorders in children and adolescents. Large rigorous RCTs are required to inform clinical care. In the meantime, clinicians must balance patient expectations with the limited evidence available.
Collapse
Affiliation(s)
- Lauren J Rice
- The University of Sydney, Faculty of Medicine and Health, Specialty of Child and Adolescent Health, Sydney, NSW, Australia.
- Sydney Children's Hospitals Network, Kids Research, Sydney, Australia.
- The University of Sydney, Faculty of Medicine and Health, Brain and Mind Centre, Sydney, NSW, Australia.
| | - Lisa Cannon
- The University of Sydney, Faculty of Medicine and Health, Specialty of Child and Adolescent Health, Sydney, NSW, Australia
- Telethon Kids Institute, Perth Children's Hospital, Perth, WA, Australia
| | - Navin Dadlani
- The University of Sydney, Faculty of Medicine and Health, Brain and Mind Centre, Sydney, NSW, Australia
| | - Melissa Mei Yin Cheung
- The University of Sydney, Faculty of Medicine and Health, Specialty of Child and Adolescent Health, Sydney, NSW, Australia
- Sydney Children's Hospitals Network, Kids Research, Sydney, Australia
| | - Stewart L Einfeld
- The University of Sydney, Faculty of Medicine and Health, Brain and Mind Centre, Sydney, NSW, Australia
| | - Daryl Efron
- Department of General Paediatrics, Health Services, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - David R Dossetor
- Sydney Children's Hospitals Network, Kids Research, Sydney, Australia
| | - Elizabeth J Elliott
- The University of Sydney, Faculty of Medicine and Health, Specialty of Child and Adolescent Health, Sydney, NSW, Australia
- Sydney Children's Hospitals Network, Kids Research, Sydney, Australia
| |
Collapse
|
8
|
Schouten M, Dalle S, Mantini D, Koppo K. Cannabidiol and brain function: current knowledge and future perspectives. Front Pharmacol 2024; 14:1328885. [PMID: 38288087 PMCID: PMC10823027 DOI: 10.3389/fphar.2023.1328885] [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: 10/27/2023] [Accepted: 12/19/2023] [Indexed: 01/31/2024] Open
Abstract
Cannabidiol (CBD) is a naturally occurring non-psychoactive cannabinoid found in Cannabis sativa, commonly known as cannabis or hemp. Although currently available CBD products do not meet the safety standards of most food safety authorities to be approved as a dietary supplement or food additive, CBD has been gaining widespread attention in recent years due to its various potential health benefits. While primarily known for its therapeutic effects in managing epileptic seizures, psychosis, anxiety, (neuropathic) pain, and inflammation, CBD's influence on brain function has also piqued the interest of researchers and individuals seeking to enhance cognitive performance. The primary objective of this review is to gather, synthesize, and consolidate scientifically proven evidence on the impact of CBD on brain function and its therapeutic significance in treating neurological and mental disorders. First, basic background information on CBD, including its biomolecular properties and mechanisms of action is presented. Next, evidence for CBD effects in the human brain is provided followed by a discussion on the potential implications of CBD as a neurotherapeutic agent. The potential effectiveness of CBD in reducing chronic pain is considered but also in reducing the symptoms of various brain disorders such as epilepsy, Alzheimer's, Huntington's and Parkinson's disease. Additionally, the implications of using CBD to manage psychiatric conditions such as psychosis, anxiety and fear, depression, and substance use disorders are explored. An overview of the beneficial effects of CBD on aspects of human behavior, such as sleep, motor control, cognition and memory, is then provided. As CBD products remain largely unregulated, it is crucial to address the ethical concerns associated with their use, including product quality, consistency, and safety. Therefore, this review discusses the need for responsible research and regulation of CBD to ensure its safety and efficacy as a therapeutic agent for brain disorders or to stimulate behavioral and cognitive abilities of healthy individuals.
Collapse
Affiliation(s)
- Moniek Schouten
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| | - Sebastiaan Dalle
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| | - Dante Mantini
- Movement Control and Neuroplasticity Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| | - Katrien Koppo
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Gamble MC, Miracle S, Williams BR, Logan RW. Sex-specific Effects of the Endocannabinoid Agonist 2-Arachidonoylglycerol on Sleep and Circadian Disruptions during Fentanyl Withdrawal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.19.572466. [PMID: 38187736 PMCID: PMC10769247 DOI: 10.1101/2023.12.19.572466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Fentanyl has become the leading driver of opioid overdoses. Cessation of opioid use represents a challenge as the experience of withdrawal drives subsequent relapse. One of the most prominent withdrawal symptoms that can contribute to opioid craving and vulnerability to relapse is sleep disruption. The endocannabinoid agonist, 2-Arachidonoylglycerol (2-AG), may promote sleep and reduce withdrawal severity; however, the effects of 2-AG on sleep disruption during opioid withdrawal have yet to be assessed. Here, we investigate the effects of 2-AG administration on sleep-wake behavior and diurnal activity in mice during withdrawal from fentanyl. Sleep-wake activity was continuously recorded before and after chronic fentanyl administration in both male and female C57BL/6J mice. Immediately following cessation of fentanyl administration, 2-AG was administered intraperitoneally to investigate the impact of endocannabinoid agonism on opioid-induced sleep disruption. Female mice maintained higher activity levels in response to chronic fentanyl than male mice. Furthermore, fentanyl increased wake and decreased sleep during the light period and inversely increased sleep and decreased wake in the dark period in both sexes. 2-AG treatment increased arousal and decreased sleep in both sexes during first 24 hrs of withdrawal. On withdrawal day 2, only female showed increased wakefulness with no changes in males, but by withdrawal day 3 male mice displayed decreased rapid-eye movement sleep during the dark period with no changes in female mice. Overall, repeated administration of fentanyl altered sleep and diurnal activity and administration of the endocannabinoid agonist, 2-AG, had sex-specific effects on fentanyl-induced sleep and diurnal changes.
Collapse
|
10
|
Gaston SA, Alhasan DM, Jones RD, Braxton Jackson W, Kesner AJ, Buxton OM, Jackson CL. Cannabis use and sleep disturbances among White, Black, and Latino adults in the United States: A cross-sectional study of National Comorbidity Survey-Replication (2001-2003) data. Sleep Health 2023; 9:587-595. [PMID: 37574376 DOI: 10.1016/j.sleh.2023.06.003] [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: 01/16/2023] [Revised: 05/03/2023] [Accepted: 06/13/2023] [Indexed: 08/15/2023]
Abstract
OBJECTIVE Research investigating cannabis use and sleep health is limited, and results are mixed. Few studies were nationally representative with racially-ethnically diverse samples or assessed potential modifiers. Our objective was to investigate cross-sectional associations between reported cannabis use and sleep disturbances by potential modifiers among non-Hispanic White, non-Hispanic Black, and Hispanic/Latino men and women in the United States. METHODS We used nationally representative National Comorbidity Survey-Replication data collected from 2001 to 2003 among 3929 adults. Poisson regression with robust variance estimated prevalence ratios (PR) and 95% confidence intervals of patterns of sleep disturbances identified through latent class analysis. Models adjusted for sociodemographic, health behavior, and clinical characteristics were stratified by race-ethnicity and by race-ethnicity along with sex/gender, and age, separately. RESULTS Over half of adults reported cannabis use (52%-ever/lifetime vs 48%-never). We identified two latent classes: multiple sleep disturbances with daytime sleepiness and no sleep disturbances with some daytime sleepiness. Prevalence of multiple sleep disturbances with daytime sleepiness was higher among participants reporting lifetime cannabis use (23% vs 20%). Associations did not vary by race-ethnicity or sex/gender. Lifetime vs never cannabis use was marginally associated with a higher prevalence of multiple sleep disturbances with daytime sleepiness only among adults aged 25-29years (PR=1.09 [95% confidence interval: 1.00-1.18]; eg, PRage 40+ years=1.00 [0.97-1.03], pinteraction=0.03). CONCLUSIONS Associations between cannabis use and sleep may vary by age. Replication with more recent data and prospective studies that investigate intersectional identities among diverse populations with objective assessments are warranted.
Collapse
Affiliation(s)
- Symielle A Gaston
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Dana M Alhasan
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Rodney D Jones
- Department of Biobehavioral Health, College of Health and Human Development, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - W Braxton Jackson
- Social & Scientific Systems, a DLH Holding Company, Durham, North Carolina, USA
| | - Andrew J Kesner
- Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Orfeu M Buxton
- Department of Biobehavioral Health, College of Health and Human Development, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Chandra L Jackson
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA; Division of Intramural Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA.
| |
Collapse
|
11
|
Belali R, Mard SA, Khoshnam SE, Bavarsad K, Sarkaki A, Farbood Y. Anandamide improves food intake and orexinergic neuronal activity in the chronic sleep deprivation induction model in rats by modulating the expression of the CB1 receptor in the lateral hypothalamus. Neuropeptides 2023; 101:102336. [PMID: 37290176 DOI: 10.1016/j.npep.2023.102336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/24/2023] [Accepted: 04/02/2023] [Indexed: 06/10/2023]
Abstract
Sleep deprivation alters orexinergic neuronal activity in the lateral hypothalamus (LH), which is the main regulator of sleep-wake, arousal, appetite, and energy regulation processes. Cannabinoid receptor (CBR) expression in this area is involved in modulating the function of orexin neurons. In this study, we investigated the effects of endocannabinoid anandamide (AEA) administration on improving food intake and appetite by modulating the activity of orexin neurons and CB1R expression after chronic sleep deprivation. Adult male Wistar rats (200-250 g) were randomly divided into three groups: control + vehicle (Control), chronic sleep deprivation + vehicle (SD), and chronic sleep deprivation +20 mg/kg AEA (SD + A). For SD induction, the rats were kept in a sleep deprivation device for 18 h (7 a.m. to 1 a.m.) daily for 21 days. Weight gain, food intake, the electrical power of orexin neurons, CB1R mRNA expression in hypothalamus, CB1R protein expression in the LH, TNF-α, IL-6, IL-4 levels and antioxidant activity in hypothalamus were measured after SD induction. Our results showed that AEA administration significantly improved food intake (p < 0.01), Electrical activity of orexin neurons (p < 0.05), CB1R expression in the hypothalamus (p < 0.05), and IL-4 levels (p < 0.05). AEA also reduced mRNA expression of OX1R and OX2R (p < 0.01 and p < 0.05 respectively), also IL-6 and TNF-α (p < 0.01) and MDA level (p < 0.05) in hypothalamic tissue. As a consequence, AEA modulates orexinergic system function and improves food intake by regulating the expression of the CB1 receptor in the LH in sleep deprived rats.
Collapse
Affiliation(s)
- Rafie Belali
- Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Ali Mard
- Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kowsar Bavarsad
- Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Sarkaki
- Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Yaghoob Farbood
- Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Basic Medical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
12
|
Xie JF, Wang LX, Ren WT, Wang C, Gao JX, Chen HL, Zhao XQ, Ren YL, Xie YP, Shao YF, Hou YP. An α-hemoglobin-derived peptide (m)VD-hemopressin (α) promotes NREM sleep via the CB 1 cannabinoid receptor. Front Pharmacol 2023; 14:1213215. [PMID: 37456761 PMCID: PMC10347404 DOI: 10.3389/fphar.2023.1213215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Hemopressin and related peptides have shown to function as the endogenous ligands or the regulator of cannabinoid receptors. The previous studies demonstrated that the endocannabinoid system played important roles in modulating several physiological functions such as sleep, olfaction, emotion, learning and memory, and reward behaviors. Mouse VD-hemopressin (α) [(m)VD-HPα], an 11-residue peptide derived from the α1 chain of hemoglobin, was recently presumed as a selective agonist of the CB1 receptor. The present study was undertaken to investigate the effects of (m)VD-HPα on the sleep-wake cycle and power spectrum of cortical EEG in freely moving rats and the potential neurons in the brain activated by (m)VD-HPα. The results showed that 20.1 nmol of (m)VD-HPα i.c.v. administration increased non-rapid eye movement (NREM) sleep in the first 2 h section accompanied by an increase in EEG delta (0.5-4 Hz) activity. The (m)VD-HPα-induced NREM sleep enhancement was due to extended episode duration instead of the episode number. In addition, the effect of (m)VD-HPα (20.1 nmol) on sleep-wake states was significantly attenuated by an antagonist of the CB1 receptor, AM251 (20 nmol, i.c.v.) but not by the CB2 receptor antagonist, AM630 (20 nmol, i.c.v.). In comparison with vehicle, (m)VD-HPα increased Fos-immunoreactive (-ir) neurons in the ventrolateral preoptic nucleus (VLPO), but reduced Fos-ir neurons in the lateral hypothalamus (LH), tuberomammillary nucleus (TMN), and locus coeruleus (LC). These findings suggest that (m)VD-HPα promotes NREM sleep via the CB1 cannabinoid receptor to probably activate VLPO GABAergic neurons, but inactivates the LH orexinergic, LC noradrenergic, and TMN histaminergic neurons.
Collapse
Affiliation(s)
- Jun-Fan Xie
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Lin-Xin Wang
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Departments of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Gansu University of Chinese Medicine, Lanzhou, China
| | - Wen-Ting Ren
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Can Wang
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Departments of Anatomy, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Jin-Xian Gao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Sleep Medicine Center of Gansu Provincial Hospital, Lanzhou, China
| | - Hai-Lin Chen
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xue-Qi Zhao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yan-Li Ren
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yu-Ping Xie
- Sleep Medicine Center of Gansu Provincial Hospital, Lanzhou, China
| | - Yu-Feng Shao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yi-Ping Hou
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
13
|
Gendy MNS, Frey BN, Van Ameringen M, Kuhathasan N, MacKillop J. Cannabidiol as a candidate pharmacotherapy for sleep disturbance in alcohol use disorder. Alcohol Alcohol 2023:7150867. [PMID: 37139966 DOI: 10.1093/alcalc/agad031] [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: 10/05/2022] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 05/05/2023] Open
Abstract
Among individuals with alcohol use disorder (AUD), it is estimated that the majority suffer from persistent sleep disturbances for which few candidate medications are available. Our aim wass to critically review the potential for cannabidiol (CBD) as a treatment for AUD-induced sleep disturbance. As context, notable side effects and abuse liability for existing medications for AUD-induced sleep disturbance reduce their clinical utility. CBD modulation of the endocannabinoid system and favorable safety profile have generated substantial interest in its potential therapeutic use for various medical conditions. A number of preclinical and clinical studies suggest promise for CBD in restoring the normal sleep-wake cycle and in enhancing sleep quality in patients diagnosed with AUD. Based on its pharmacology and the existing literature, albeit primarily preclinical and indirect, CBD is a credible candidate to address alcohol-induced sleep disturbance. Well-designed RCTs will be necessary to test its potential in managing this challenging feature of AUD.
Collapse
Affiliation(s)
- Marie N S Gendy
- Peter Boris Centre for Addictions Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, ON L8P 3R2, Canada
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON L8N 3K7, Canada
| | - Benicio N Frey
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON L8N 3K7, Canada
- Women's Health Concerns Clinic at St. Joseph's Healthcare Hamilton, Hamilton, ON L8N 3K7, Canada
- Mood Disorders Program, St. Joseph's Healthcare Hamilton, Hamilton, ON L8N 3K7, Canada
| | - Michael Van Ameringen
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON L8N 3K7, Canada
| | - Nirushi Kuhathasan
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON L8N 3K7, Canada
- Mood Disorders Program, St. Joseph's Healthcare Hamilton, Hamilton, ON L8N 3K7, Canada
- Michael G. DeGroote Centre for Medicinal Cannabis Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, ON L8S 4K1, Canada
| | - James MacKillop
- Peter Boris Centre for Addictions Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, ON L8P 3R2, Canada
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON L8N 3K7, Canada
- Women's Health Concerns Clinic at St. Joseph's Healthcare Hamilton, Hamilton, ON L8N 3K7, Canada
- Mood Disorders Program, St. Joseph's Healthcare Hamilton, Hamilton, ON L8N 3K7, Canada
- Michael G. DeGroote Centre for Medicinal Cannabis Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
14
|
Nakagawa Y, Yamada S. Alterations in Brain Neural Network and Stress System in Atopic Dermatitis: Novel Therapeutic Interventions. J Pharmacol Exp Ther 2023; 385:78-87. [PMID: 36828629 DOI: 10.1124/jpet.122.001482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/02/2023] [Accepted: 02/03/2023] [Indexed: 02/26/2023] Open
Abstract
Atopic dermatitis is a common chronic inflammatory skin disease, with most cases experiencing skin barrier dysfunction and enhanced allergen entry, accompanied by cytokine production which evokes predominantly type-2-skewed immune responses, itch, and scratching behavior. Although intense itch and excessive scratching behavior affect progression of skin lesions, it is unclear what causes them. Data suggest that scratching behavior stimulates brain dopaminergic reward and habit learning systems, strengthening habitual scratching behavior, while nocturnal scratching behavior presumably increases locus coeruleus-noradrenergic system activity, prompting sleep disturbances. At the early stage of atopic dermatitis, increased cortisol levels, due to hypothalamic-pituitary-adrenal axis overactivation caused by such system stimulation, can induce dorsolateral prefrontal cortex disturbance with reinforcement of habitual scratching behavior and may aggravate type-2-skewed immune responses in the skin. During the later phases, whereas blunted hypothalamic-pituitary-adrenal axis function and the shift of type-2-dominated to type-1-co-dominated inflammation are induced, noradrenergic system overactivation-associated dorsolateral prefrontal cortex disruption is ongoing and responsible for itch cognitive distortion to catastrophize about itch, which leads to a vicious spiral along with habitual scratching behavior and skin lesions. Data are presented in this review indicating that while skin immune system dysfunction initiates pathologic changes in atopic dermatitis, brain neural network and stress system alterations can promote the progression of this condition. It is also suggested that cognitive distortion contributes to pathology in atopic dermatitis as with some psychiatric disorders and chronic pain. The proposed mechanistic model could lead to development of novel medications for slowing or terminating the relentless progression of this disorder. SIGNIFICANCE STATEMENT: Although conventional pharmacological interventions focusing on skin homeostasis and itch occurrence significantly attenuate clinical signs in atopic dermatitis patients, achievement of 100% improvement is less than 40% in several double-blind, randomized, placebo-controlled trials. Our model predicts that itch cognitive distortion, due to dorsolateral prefrontal cortex disturbance, can significantly contribute to the progression of atopic dermatitis and that agents capable of improving brain neural network, stress system, and skin homeostasis may be effective as interventions in the treatment of this condition.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| | - Shizuo Yamada
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| |
Collapse
|
15
|
Coordinated Regulation of CB1 Cannabinoid Receptors and Anandamide Metabolism Stabilizes Network Activity during Homeostatic Downscaling. eNeuro 2022; 9:ENEURO.0276-22.2022. [PMID: 36316118 PMCID: PMC9663203 DOI: 10.1523/eneuro.0276-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 12/24/2022] Open
Abstract
Neurons express overlapping homeostatic mechanisms to regulate synaptic function and network properties in response to perturbations of neuronal activity. Endocannabinoids (eCBs) are bioactive lipids synthesized in the postsynaptic compartments to regulate synaptic transmission, plasticity, and neuronal excitability primarily through retrograde activation of presynaptic cannabinoid receptor type 1 (CB1). The eCB system is well situated to regulate neuronal network properties and coordinate presynaptic and postsynaptic activity. However, the role of the eCB system in homeostatic adaptations to neuronal hyperactivity is unknown. To address this issue, we used Western blotting and targeted lipidomics to measure adaptations in eCB system to bicuculline (BCC)-induced chronic hyperexcitation in mature cultured rat cortical neurons, and used multielectrode array (MEA) recording and live-cell imaging of glutamate dynamics to test the effects of pharmacological manipulations of eCB on network activities. We show that BCC-induced chronic hyperexcitation triggers homeostatic downscaling and a coordinated adaptation to enhance tonic eCB signaling. Hyperexcitation triggers first the downregulation of fatty acid amide hydrolase (FAAH), the lipase that degrades the eCB anandamide, then an accumulation of anandamide and related metabolites, and finally a delayed upregulation of surface and total CB1. Additionally, we show that BCC-induced downregulation of surface AMPA-type glutamate receptors (AMPARs) and upregulation of CB1 occur through independent mechanisms. Finally, we show that endocannabinoids support baseline network activities before and after downscaling and is engaged to suppress network activity during adaptation to hyperexcitation. We discuss the implications of our findings in the context of downscaling and homeostatic regulation of in vitro oscillatory network activities.
Collapse
|
16
|
Martin SC, Gay SM, Armstrong ML, Pazhayam NM, Reisdorph N, Diering GH. Tonic endocannabinoid signaling supports sleep through development in both sexes. Sleep 2022; 45:6565640. [PMID: 35395682 PMCID: PMC9366650 DOI: 10.1093/sleep/zsac083] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Sleep is an essential behavior that supports brain function and cognition throughout life, in part by acting on neuronal synapses. The synaptic signaling pathways that mediate the restorative benefits of sleep are not fully understood, particularly in the context of development. Endocannabinoids (eCBs) including 2-arachidonyl glycerol (2-AG) and anandamide (AEA), are bioactive lipids that activate cannabinoid receptor, CB1, to regulate synaptic transmission and mediate cognitive functions and many behaviors, including sleep. We used targeted mass spectrometry to measure changes in forebrain synaptic eCBs during the sleep/wake cycle in juvenile and adolescent mice of both sexes. We find that eCBs lack a daily rhythm in juvenile mice, while in adolescents AEA and related oleoyl ethanolamide are increased during the sleep phase in a circadian manner. Next, we manipulated the eCB system using selective pharmacology and measured the effects on sleep behavior in developing and adult mice of both sexes using a noninvasive piezoelectric home-cage recording apparatus. Enhancement of eCB signaling through inhibition of 2-AG or AEA degradation, increased dark-phase sleep amount and bout length in developing and adult males, but not in females. Inhibition of CB1 by injection of the antagonist AM251 reduced sleep time and caused sleep fragmentation in developing and adult males and females. Our data suggest that males are more sensitive to the sleep-promoting effects of enhanced eCBs but that tonic eCB signaling supports sleep behavior through multiple stages of development in both sexes. This work informs the further development of cannabinoid-based therapeutics for sleep disruption.
Collapse
Affiliation(s)
- Shenée C Martin
- Department of Cell Biology and Physiology and the UNC Neuroscience Center, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - Sean M Gay
- Department of Cell Biology and Physiology and the UNC Neuroscience Center, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - Michael L Armstrong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, CO , USA
| | - Nila M Pazhayam
- Department of Cell Biology and Physiology and the UNC Neuroscience Center, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, CO , USA
| | - Graham H Diering
- Department of Cell Biology and Physiology and the UNC Neuroscience Center, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
- Carolina Institute for Developmental Disabilities , Carrboro, NC , USA
| |
Collapse
|
17
|
Kesner AJ, Mateo Y, Abrahao KP, Ramos-Maciel S, Pava MJ, Gracias AL, Paulsen RT, Carlson HB, Lovinger DM. Changes in striatal dopamine release, sleep, and behavior during spontaneous Δ-9-tetrahydrocannabinol abstinence in male and female mice. Neuropsychopharmacology 2022; 47:1537-1549. [PMID: 35478010 PMCID: PMC9205922 DOI: 10.1038/s41386-022-01326-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/16/2022] [Accepted: 04/11/2022] [Indexed: 11/09/2022]
Abstract
Withdrawal symptoms are observed upon cessation of cannabis use in humans. Although animal studies have examined withdrawal symptoms following exposure to delta-9-tetrahydrocannabinol (THC), difficulties in obtaining objective measures of spontaneous withdrawal using paradigms that mimic cessation of use in humans have slowed research. The neuromodulator dopamine (DA) is affected by chronic THC treatment and plays a role in many behaviors related to human THC withdrawal symptoms. These symptoms include sleep disturbances that often drive relapse, and emotional behaviors like irritability and anhedonia. We examined THC withdrawal-induced changes in striatal DA release and the extent to which sleep disruption and behavioral maladaptation manifest during abstinence in a mouse model of chronic THC exposure. Using a THC treatment regimen known to produce tolerance, we measured electrically elicited DA release in acute brain slices from different striatal subregions during early and late THC abstinence. Long-term polysomnographic recordings from mice were used to assess vigilance state and sleep architecture before, during, and after THC treatment. We additionally assessed how behaviors that model human withdrawal symptoms are altered by chronic THC treatment in early and late abstinence. We detected altered striatal DA release, sleep disturbances that mimic clinical observations, and behavioral maladaptation in mice following tolerance to THC. Altered striatal DA release, sleep, and affect-related behaviors associated with spontaneous THC abstinence were more consistently observed in male mice. These findings provide a foundation for preclinical study of directly translatable non-precipitated THC withdrawal symptoms and the neural mechanisms that affect them.
Collapse
Affiliation(s)
- Andrew J Kesner
- National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Yolanda Mateo
- National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Karina P Abrahao
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Campus São Paulo, São Paulo, SP, Brazil
| | - Stephanie Ramos-Maciel
- National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | | | - Alexa L Gracias
- National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Riley T Paulsen
- National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Hartley B Carlson
- National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - David M Lovinger
- National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA.
| |
Collapse
|
18
|
Ramon-Duaso C, Conde-Moro AR, Busquets-Garcia A. Astroglial cannabinoid signaling and behavior. Glia 2022; 71:60-70. [PMID: 35293647 DOI: 10.1002/glia.24171] [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: 01/13/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 11/11/2022]
Abstract
In neuroscience, the explosion of innovative and advanced technical accomplishments is fundamental to understanding brain functioning. For example, the possibility to distinguish glial and neuronal activities at the synaptic level and/or the appearance of new genetic tools to specifically monitor and manipulate astroglial functions revealed that astrocytes are involved in several facets of behavioral control. In this sense, the discovery of functional presence of type-1 cannabinoid receptors in astrocytes has led to identify important behavioral responses mediated by this specific pool of cannabinoid receptors. Thus, astroglial type-1 cannabinoid receptors are in the perfect place to play a role in a complex scenario in which astrocytes sense neuronal activity, release gliotransmitters and modulate the activity of other neurons, ultimately controlling behavioral responses. In this review, we will describe the known behavioral implications of astroglial cannabinoid signaling and highlight exciting unexplored research avenues on how astroglial cannabinoid signaling could affect behavior.
Collapse
Affiliation(s)
- Carla Ramon-Duaso
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Programme, IMIM Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Ana Rocio Conde-Moro
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Programme, IMIM Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Arnau Busquets-Garcia
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Programme, IMIM Hospital del Mar Medical Research Institute, Barcelona, Spain
| |
Collapse
|
19
|
Peng J, Fan M, An C, Ni F, Huang W, Luo J. A narrative review of molecular mechanism and therapeutic effect of Cannabidiol (CBD). Basic Clin Pharmacol Toxicol 2022; 130:439-456. [PMID: 35083862 DOI: 10.1111/bcpt.13710] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 11/28/2022]
Abstract
Cannabidiol (CBD) is an abundant non-psychoactive phytocannabinoid in Cannabis extracts which has high affinity on a series of receptors, including type 1 cannabinoid receptor (CB1), type 2 cannabinoid receptor (CB2), GPR55, transient receptor potential vanilloid (TRPV), and peroxisome proliferator-activated receptor gamma (PPARγ). By modulating the activities of these receptors, CBD exhibits multiple therapeutic effects, including neuroprotective, antiepileptic, anxiolytic, antipsychotic, anti-inflammatory, analgesic and anti-cancer properties. CBD could also be applied to treat or prevent COVID-19 and its complications. Here, we provide a narrative review of CBD's applications in human diseases: from mechanism of action to clinical trials.
Collapse
Affiliation(s)
- Jiangling Peng
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Mingjie Fan
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Chelsea An
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang, China
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | | |
Collapse
|
20
|
Possible actions of cannabidiol in obsessive-compulsive disorder by targeting the WNT/β-catenin pathway. Mol Psychiatry 2022; 27:230-248. [PMID: 33837269 DOI: 10.1038/s41380-021-01086-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/13/2021] [Accepted: 03/26/2021] [Indexed: 02/02/2023]
Abstract
Obsessive-compulsive disorder (OCD) is a neuropsychiatric disorder characterized by recurrent and distinctive obsessions and/or compulsions. The etiologies remain unclear. Recent findings have shown that oxidative stress, inflammation, and glutamatergic pathways play key roles in the causes of OCD. However, first-line therapies include cognitive-behavioral therapy but only 40% of the patients respond to this first-line therapy. Research for new treatment is mandatory. This review focuses on the potential effects of cannabidiol (CBD), as a potential therapeutic strategy, on OCD and some of the presumed mechanisms by which CBD provides its benefit properties. CBD medication downregulates GSK-3β, the main inhibitor of the WNT/β-catenin pathway. The activation of the WNT/β-catenin could be associated with the control of oxidative stress, inflammation, and glutamatergic pathway and circadian rhythms dysregulation in OCD. Future prospective clinical trials could focus on CBD and its different and multiple interactions in OCD.
Collapse
|
21
|
Murillo-Rodríguez E. The Endocannabinoid System as Prognostic Biomarker of the Obstructive Sleep Apnea Morbidity in COVID-19-Recovered Individuals. SLEEP AND VIGILANCE 2021; 5:205-211. [PMID: 34604693 PMCID: PMC8475390 DOI: 10.1007/s41782-021-00169-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/28/2021] [Accepted: 09/16/2021] [Indexed: 11/29/2022]
Abstract
The endocannabinoid system is a neurobiological signaling network that is present in the human biological systems, including the brain. This neurobiological system comprises cannabinoid receptors, endogenous ligands, as well as enzymatic synthesis, degradation and transport of endocannabinoids and has been suggested as a modulator of multiple physiological processes, including the sleep–wake cycle. On the other hand, the COVID-19 pandemic, originated by the novel coronavirus SARS-CoV-2, has caused global catastrophes in economic, social, and health spheres. COVID-19 is a multi-organ disease with a broad spectrum of health complications, such as respiratory infections leading to respiratory-related symptoms and disorders. The development, approval, and application of vaccines against SARS-CoV-2 is ongoing; however, there are increasing reports of prolonged effects after COVID-19 infection, including respiratory and neurological sequelae. Here, I provide a comprehensive review of the current literature on the endocannabinoid system and their role in sleep modulation. Whilst I discuss relevant considerations for the high risk for developing sleep disorders related to respiratory failures, such as obstructive sleep apnea (OSA) in recovered COVID-19-infected subjects. Finally, I propose a framework that integrates the analysis of the components of the endocannabinoid system as prognostic biomarkers of the likely OSA after COVID-19 infection.
Collapse
|
22
|
Li J, Carvajal R, Bruner L, Kaminski NE. The current understanding of the benefits, safety, and regulation of cannabidiol in consumer products. Food Chem Toxicol 2021; 157:112600. [PMID: 34626752 DOI: 10.1016/j.fct.2021.112600] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 01/04/2023]
Abstract
The popularity of cannabidiol (CBD) in consumer products is soaring as consumers are using CBD for general health and well-being as well as to seek relief from ailments especially pain, inflammation, anxiety, depression, and sleep disorders. However, there are limited data currently in the public domain that provide support for these benefits. By contrast, a significant amount of safety evaluation data for CBD have been obtained recently from pre-clinical and clinical studies of the CBD therapeutic Epidiolex®. Yet some key data gaps concerning the safe use of CBD still remain. Furthermore, current regulations on CBD use in consumer products remain uncertain and often conflict between the state and federal level. In light of the rapidly expanding popularity of CBD-related products in the marketplace, here we review the current understanding of the benefits, safety, and regulations surrounding CBD in consumer products. This review does not advocate for or against the use of CBD in consumer products. Rather this review seeks to assess the state-of-the-science on the health effects and safety of CBD, to identify critical knowledge gaps for future studies, and to raise the awareness of the current regulations that govern CBD use in consumer products.
Collapse
Affiliation(s)
- Jinpeng Li
- Center for Research on Ingredient Safety, Michigan State University, East Lansing, MI, 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | | | - Leon Bruner
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Norbert E Kaminski
- Center for Research on Ingredient Safety, Michigan State University, East Lansing, MI, 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
23
|
Soundararajan S, Kazmi N, Brooks AT, Krumlauf M, Schwandt ML, George DT, Hodgkinson CA, Wallen GR, Ramchandani VA. FAAH and CNR1 Polymorphisms in the Endocannabinoid System and Alcohol-Related Sleep Quality. Front Psychiatry 2021; 12:712178. [PMID: 34566715 PMCID: PMC8458733 DOI: 10.3389/fpsyt.2021.712178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/09/2021] [Indexed: 12/02/2022] Open
Abstract
Sleep disturbances are common among individuals with alcohol use disorder (AUD) and may not resolve completely with short-term abstinence from alcohol, potentially contributing to relapse to drinking. The endocannabinoid system (ECS) is associated with both sleep and alcohol consumption, and genetic variation in the ECS may underlie sleep-related phenotypes among individuals with AUD. In this study, we explored the influence of genetic variants in the ECS (Cannabinoid receptor 1/CNR1: rs806368, rs1049353, rs6454674, rs2180619, and Fatty Acid Amide Hydrolase/FAAH rs324420) on sleep quality in individuals with AUD (N = 497) and controls without AUD (N = 389). We assessed subjective sleep quality (from the Pittsburgh Sleep Quality Index/PSQI) for both groups at baseline and objective sleep efficiency and duration (using actigraphy) in a subset of individuals with AUD at baseline and after 4 weeks of inpatient treatment. We observed a dose-dependent relationship between alcohol consumption and sleep quality in both AUD and control groups. Sleep disturbance, a subscale measure in PSQI, differed significantly among CNR1 rs6454674 genotypes in both AUD (p = 0.015) and controls (p = 0.016). Only among controls, neuroticism personality scores mediated the relationship between genotype and sleep disturbance. Objective sleep measures (sleep efficiency, wake bouts and wake after sleep onset), differed significantly by CNR1 rs806368 genotype, both at baseline (p = 0.023, 0.029, 0.015, respectively) and at follow-up (p = 0.004, p = 0.006, p = 0.007, respectively), and by FAAH genotype for actigraphy recorded sleep duration at follow-up (p = 0.018). These relationships suggest a significant role of the ECS in alcohol-related sleep phenotypes.
Collapse
Affiliation(s)
- Soundarya Soundararajan
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Narjis Kazmi
- National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Alyssa T. Brooks
- National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Michael Krumlauf
- National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Melanie L. Schwandt
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - David T. George
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Colin A. Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Gwenyth R. Wallen
- National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Vijay A. Ramchandani
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
24
|
Vaseghi S, Arjmandi-Rad S, Nasehi M, Zarrindast MR. Cannabinoids and sleep-wake cycle: The potential role of serotonin. Behav Brain Res 2021; 412:113440. [PMID: 34216647 DOI: 10.1016/j.bbr.2021.113440] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/28/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
Cannabis sativa (Marijuana) has a long history as a medicinal plant and Δ9-tetrahydrocannabinol (Δ9-THC) is the most active component in this plant. Cannabinoids are interesting compounds with various modulatory effects on physiological processes and cognitive functions. The use of cannabinoids is a double-edged sword, because they induce both adverse and therapeutic properties. One of the most important roles of cannabinoids is modulating sleep-wake cycle. Sleep, its cycle, and its mechanism are highly unknown. Also, the effects of cannabinoids on sleep-wake cycle are so inconsistent. Thus, understanding the role of cannabinoids in modulating sleep-wake cycle is a critical scientific goal. Cannabinoids interact with many neurotransmitter systems. In this review article, we chose serotonin due to its important role in regulating sleep-wake cycle. We found that the interaction between cannabinoids and serotonergic signaling especially in the dorsal raphe is extensive, unknown, and controversial.
Collapse
Affiliation(s)
- Salar Vaseghi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran.
| | - Shirin Arjmandi-Rad
- Institute for Cognitive & Brain Sciences, Shahid Beheshti University, Tehran, Iran
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran; Department of Pharmacology School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Morris G, Walder K, Kloiber S, Amminger P, Berk M, Bortolasci CC, Maes M, Puri BK, Carvalho AF. The endocannabinoidome in neuropsychiatry: Opportunities and potential risks. Pharmacol Res 2021; 170:105729. [PMID: 34119623 DOI: 10.1016/j.phrs.2021.105729] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023]
Abstract
The endocannabinoid system (ECS) comprises two cognate endocannabinoid receptors referred to as CB1R and CB2R. ECS dysregulation is apparent in neurodegenerative/neuro-psychiatric disorders including but not limited to schizophrenia, major depressive disorder and potentially bipolar disorder. The aim of this paper is to review mechanisms whereby both receptors may interact with neuro-immune and neuro-oxidative pathways, which play a pathophysiological role in these disorders. CB1R is located in the presynaptic terminals of GABAergic, glutamatergic, cholinergic, noradrenergic and serotonergic neurons where it regulates the retrograde suppression of neurotransmission. CB1R plays a key role in long-term depression, and, to a lesser extent, long-term potentiation, thereby modulating synaptic transmission and mediating learning and memory. Optimal CB1R activity plays an essential neuroprotective role by providing a defense against the development of glutamate-mediated excitotoxicity, which is achieved, at least in part, by impeding AMPA-mediated increase in intracellular calcium overload and oxidative stress. Moreover, CB1R activity enables optimal neuron-glial communication and the function of the neurovascular unit. CB2R receptors are detected in peripheral immune cells and also in central nervous system regions including the striatum, basal ganglia, frontal cortex, hippocampus, amygdala as well as the ventral tegmental area. CB2R upregulation inhibits the presynaptic release of glutamate in several brain regions. CB2R activation also decreases neuroinflammation partly by mediating the transition from a predominantly neurotoxic "M1" microglial phenotype to a more neuroprotective "M2" phenotype. CB1R and CB2R are thus novel drug targets for the treatment of neuro-immune and neuro-oxidative disorders including schizophrenia and affective disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Stefan Kloiber
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 33 Ursula Franklin Street, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Paul Amminger
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| |
Collapse
|
26
|
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
|
27
|
Puskar P, Sengupta T, Sharma B, Nath SS, Mallick H, Akhtar N. Changes in sleep-wake cycle after microinjection of agonist and antagonist of endocannabinoid receptors at the medial septum of rats. Physiol Behav 2021; 237:113448. [PMID: 33957148 DOI: 10.1016/j.physbeh.2021.113448] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 04/18/2021] [Accepted: 04/29/2021] [Indexed: 01/06/2023]
Abstract
The role of medial septum in the genesis of slow-wave sleep and the inhibition of rapid eye movement sleep has been established using neurotoxic lesion and chemical stimulation of the medial septum. Intracerebroventricular injection of endocannabinoids (anandamide) decreases wake and increases slow-wave and rapid eye movement sleep in rats. Central cannabinoid (CB1) receptors are localized in the rat medial septum; however, the role of cannabinoid receptors at the medial septum on the regulation of sleep-wakefulness in rats lacks evidence. In this study, we have examined the changes in sleep architecture of 21 male Wistar rats, divided into three groups. Initially, 6 rats were used for dose standardization. Subsequently, one group (n = 6) was microinjected with CB1 receptor agonist, R-(+)-WIN 55,212-2 mesylate salt, the second group (n = 6) received microinjection of CB1 receptor antagonist LY 320,135, and the third group (n = 5) was microinjected with the vehicle, DMSO at the medial septum using stereotaxy. The sleep-wake cycle was recorded using electroencephalogram, electro-oculogram, and electromyogram. Microinjection of CB1 receptor agonist at the medial septum decreased slow-wave sleep and increased total sleep time. The increase in total sleep time was due to an increased percentage of rapid eye movement sleep. After the third and fourth hour of CB1 receptor antagonist microinjection at the medial septum, slow-wave sleep decreased when compared to vehicle injection, while rapid eye movement sleep decreased compared to baseline. We conclude that the endocannabinoid system at the septal nucleus acts through CB1 receptors to increase rapid eye movement sleep in rats.
Collapse
Affiliation(s)
- Preeti Puskar
- All India Institute of Medical Sciences, New Delhi-110029, India.
| | - Trina Sengupta
- All India Institute of Medical Sciences, Jodhpur-342005, India.
| | - Binney Sharma
- All India Institute of Medical Sciences, New Delhi-110029, India.
| | - Sriji S Nath
- All India Institute of Medical Sciences, New Delhi-110029, India.
| | - Hrudananda Mallick
- Department of Physiology, Faculty of Medicine & Health Sciences, SGT University, Gurgaon, Haryana- 122505.
| | - Nasreen Akhtar
- All India Institute of Medical Sciences, New Delhi-110029, India.
| |
Collapse
|
28
|
The quality and duration of sleep are related to hedonic hunger: a cross-sectional study in university students. Sleep Biol Rhythms 2021. [DOI: 10.1007/s41105-020-00303-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
29
|
Kaul M, Zee PC, Sahni AS. Effects of Cannabinoids on Sleep and their Therapeutic Potential for Sleep Disorders. Neurotherapeutics 2021; 18:217-227. [PMID: 33580483 PMCID: PMC8116407 DOI: 10.1007/s13311-021-01013-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2021] [Indexed: 12/19/2022] Open
Abstract
The recent trend for legalization of medicinal cannabis and cannabinoid-containing products, together with their soporific effects, has led to a surge of interest of their potential therapeutic role in the management of some common sleep disorders, such as insomnia, sleep disordered breathing, and restless legs syndrome, and less common disorders such as narcolepsy and parasomnias. Although much of the pre-clinical and clinical data were derived from studies with relatively small sample sizes and limited by biases in assessment, and in clinical trials lack of allocation concealment, as a whole, the results indicate a potential therapeutic role for cannabinoids in the management of some sleep disorders. Clinical trials are underway for insomnia and obstructive sleep apnea management, but there remains a substantial need for rigorous large multi-center studies to assess the dose, efficacy, and safety of the various types of cannabinoids on sleep disorders. This review aims to summarize the modulatory effects of cannabinoids on sleep physiology and provide a critical evaluation of the research on their potential therapeutic benefit in various sleep disorders.
Collapse
Affiliation(s)
- Malvika Kaul
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, 909 S Wolcott Ave, Room 3135 (MC 719), Chicago, IL, 60612, USA
| | - Phyllis C Zee
- Department of Neurology, Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, 305 E. Chicago Ave., Chicago, IL, 60611, USA
| | - Ashima S Sahni
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, 909 S Wolcott Ave, Room 3135 (MC 719), Chicago, IL, 60612, USA.
| |
Collapse
|
30
|
Effects of Cannabinoid Agonists and Antagonists on Sleep in Laboratory Animals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1297:97-109. [PMID: 33537939 DOI: 10.1007/978-3-030-61663-2_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The cannabinoids are a family of chemical compounds that can be either synthesized or naturally derived. These compounds have been shown to modulate a wide variety of biological processes. In this chapter, the studies detailing the effects of cannabinoids on sleep in laboratory animals are reviewed. Both exogenous and endogenous cannabinoids generally appear to decrease wakefulness and alter rapid eye movement (REM) and non-REM sleep in animal models. In addition, cannabinoids potentiate the effects of sedative-hypnotic drugs. However, the individual contributions of each cannabinoid on sleep processes is more nuanced and may depend on the site of action in the central nervous system. Many studies investigating the mechanism of cannabinoid effects on sleep suggest that the effects of cannabinoids on sleep are mediated via cannabinoid receptors; however, some evidence suggests that some sleep effects may be elicited via non-cannabinoid receptor-dependent mechanisms. More research is necessary to fully elucidate the role of each compound in modulating sleep processes.
Collapse
|
31
|
Blood endocannabinoid levels in patients with panic disorder. Psychoneuroendocrinology 2020; 122:104905. [PMID: 33091759 DOI: 10.1016/j.psyneuen.2020.104905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND The development and maintenance of anxiety disorders is not fully understood. There is consensus in the literature that in addition to genetic factors, social, psychological and neurobiological factors are of crucial importance. The present exploratory study investigates the influence of the endocannabinoids (EC) and related N-acylethanolamines (NA) on the maintenance of panic disorder (PD). METHODS A total of n = 36 PD and n = 26 healthy controls (HC) were included in the study. Baseline characteristics showed no differences between the two groups. The participants were exposed to the Trier Social Stress Test (TSST) for reliable laboratory stress induction. Blood samples were taken during the TSST by an intravenous catheter to examine the endocannabinoid (EC) stress response. Repeated measures ANOVA was conducted to test for main effects of time and group as well as the respective interaction. RESULTS Participants with PD consistently had significantly higher EC and NA blood levels than HC. The consistently high EC and NA levels barely showed any reactivity as indicated by a lack of statistical variance. In line with these findings no reaction to the psychosocial stressor TSST could be detected. CONCLUSION Our main results show significant differences in EC concentrations between participants with PD and HC. These findings suggest that an imbalance in the ECS contributes to the maintenance of PD. Increased endocannabinoid levels may have important implications for organic diseases such as cardiovascular disorders. The limitations of the study as well as implications for further investigations are discussed.
Collapse
|
32
|
Girard F, von Siebenthal M, Davis FP, Celio MR. Gene expression analysis in the mouse brainstem identifies Cart and Nesfatin as neuropeptides coexpressed in the Calbindin-positive neurons of the Nucleus papilio. Sleep 2020; 43:5826369. [PMID: 32343818 PMCID: PMC7658639 DOI: 10.1093/sleep/zsaa085] [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: 12/11/2019] [Revised: 04/08/2020] [Indexed: 11/17/2022] Open
Abstract
Study Objectives: The brainstem contains several neuronal populations, heterogeneous in terms of neurotransmitter/neuropeptide content, which are important for controlling various aspects of the rapid eye movement (REM) phase of sleep. Among these populations are the Calbindin (Calb)-immunoreactive NPCalb neurons, located in the Nucleus papilio, within the dorsal paragigantocellular nucleus (DPGi), and recently shown to control eye movement during the REM phase of sleep. Methods: We performed in-depth data mining of the in situ hybridization data collected at the Allen Brain Atlas, in order to identify potentially interesting genes expressed in this brainstem nucleus. Our attention focused on genes encoding neuropeptides, including Cart (Cocaine and Amphetamine Regulated Transcripts) and Nesfatin 1. Results: While nesfatin 1 appeared ubiquitously expressed in this Calb-positive neuronal population, Cart was coexpressed in only a subset of these glutamatergic NPCalb neurons. Furthermore, an REM sleep deprivation and rebound assay performed with mice revealed that the Cart-positive neuronal population within the DPGi was activated during REM sleep (as measured by c-fos immunoreactivity), suggesting a role of this neuropeptide in regulating some aspects of REM sleep. Conclusions: The assembled information could afford functional clues to investigators, conducive to further experimental pursuits.
Collapse
Affiliation(s)
- Franck Girard
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
| | | | - Fred P Davis
- Janelia-Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - Marco R Celio
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
33
|
Bedse G, Hill MN, Patel S. 2-Arachidonoylglycerol Modulation of Anxiety and Stress Adaptation: From Grass Roots to Novel Therapeutics. Biol Psychiatry 2020; 88:520-530. [PMID: 32197779 PMCID: PMC7486996 DOI: 10.1016/j.biopsych.2020.01.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/31/2019] [Accepted: 01/18/2020] [Indexed: 01/13/2023]
Abstract
Over the past decade there has been a surge of interest in the development of endocannabinoid-based therapeutic approaches for the treatment of diverse neuropsychiatric conditions. Although initial preclinical and clinical development efforts focused on pharmacological inhibition of fatty acid amide hydrolase to elevate levels of the endocannabinoid anandamide, more recent efforts have focused on inhibition of monoacylglycerol lipase (MAGL) to enhance signaling of the most abundant and efficacious endocannabinoid ligand, 2-arachidonoylglycerol (2-AG). We review the biochemistry and physiology of 2-AG signaling and preclinical evidence supporting a role for this system in the regulation of anxiety-related outcomes and stress adaptation. We review preclinical evidence supporting MAGL inhibition for the treatment of affective, trauma-related, and stress-related disorders; describe the current state of MAGL inhibitor drug development; and discuss biological factors that could affect MAGL inhibitor efficacy. Issues related to the clinical advancement of MAGL inhibitors are also discussed. We are cautiously optimistic, as the field of MAGL inhibitor development transitions from preclinical to clinical and theoretical to practical, that pharmacological 2-AG augmentation could represent a mechanistically novel therapeutic approach for the treatment of affective and stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gaurav Bedse
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Vanderbilt Center for Addiction Research, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mathew N Hill
- Department of Cell Biology, Hotchkiss Brain Institute and Mathison Center for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Anatomy and Psychiatry, Hotchkiss Brain Institute and Mathison Center for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sachin Patel
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Vanderbilt Center for Addiction Research, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
34
|
Peball M, Krismer F, Knaus H, Djamshidian A, Werkmann M, Carbone F, Ellmerer P, Heim B, Marini K, Valent D, Goebel G, Ulmer H, Stockner H, Wenning GK, Stolz R, Krejcy K, Poewe W, Seppi K. Non-Motor Symptoms in Parkinson's Disease are Reduced by Nabilone. Ann Neurol 2020; 88:712-722. [PMID: 32757413 PMCID: PMC7540547 DOI: 10.1002/ana.25864] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/24/2020] [Accepted: 07/26/2020] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The objective of this study was to assess the efficacy and safety of nabilone, a synthetic tetrahydrocannabinol analogue, as a treatment for non-motor symptoms (NMS) in Parkinson's disease (PD). METHODS This was a phase II placebo-controlled, double-blind, parallel-group, enriched enrollment randomized withdrawal trial conducted at the Medical University Innsbruck. A random sample of 47 patients with PD with stable motor disease and disturbing NMS defined by a score of ≥4 points on the Movement Disorder Society - Unified PD Rating Scale-I (MDS-UPDRS-I) underwent open-label nabilone titration (0.25 mg once daily to 1 mg twice daily, phase I). Responders were randomized 1:1 to continue with nabilone or switch to placebo for 4 weeks (phase II). The primary efficacy criterion was the change of the MDS-UPDRS-I between randomization and week 4. Safety was analyzed in all patients who received at least one nabilone dose. RESULTS Between October 2017 and July 2019, 19 patients received either nabilone (median dose = 0.75 mg) or placebo. At week 4, mean change of the MDS-UPDRS-I was 2.63 (95% confidence interval [CI] 1.53 to 3.74, p = 0.002, effect size = 1.15) in the placebo versus 1.00 (95% CI -0.16 to 2.16, p = 0.280, effect size = 0.42) in the nabilone-group (difference: 1.63, 95% CI 0.09 to 3.18, p = 0.030, effect size = 0.66). Seventy-seven percent of patients had adverse events (AEs) during open-label titration, most of them were transient. In the double-blind phase, similar proportions of patients in each group had AEs (42% in the placebo group and 32% in the nabilone group). There were no serious AEs. INTERPRETATION Our results highlight the potential efficacy of nabilone for patients with PD with disturbing NMS, which appears to be driven by positive effects on anxious mood and night-time sleep problems. TRIAL REGISTRY ClinicalTrials.gov (NCT03769896) and EudraCT (2017-000192-86). ANN NEUROL 2020;88:712-722.
Collapse
Affiliation(s)
- Marina Peball
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | - Florian Krismer
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | - Hans‐Günther Knaus
- Department for Medical Genetics, Molecular, and Clinical PharmacologyInnsbruck Medical UniversityInnsbruckAustria
| | | | - Mario Werkmann
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | - Federico Carbone
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | - Philipp Ellmerer
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | - Beatrice Heim
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | - Kathrin Marini
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | - Dora Valent
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | - Georg Goebel
- Department of Medical Statistics, Informatics, and Health EconomicsInnsbruck Medical UniversityInnsbruckAustria
| | - Hanno Ulmer
- Department of Medical Statistics, Informatics, and Health EconomicsInnsbruck Medical UniversityInnsbruckAustria
| | - Heike Stockner
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | | | - Raphaela Stolz
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | | | - Werner Poewe
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | - Klaus Seppi
- Department of NeurologyInnsbruck Medical UniversityInnsbruckAustria
| | | |
Collapse
|
35
|
Suraev AS, Marshall NS, Vandrey R, McCartney D, Benson MJ, McGregor IS, Grunstein RR, Hoyos CM. Cannabinoid therapies in the management of sleep disorders: A systematic review of preclinical and clinical studies. Sleep Med Rev 2020; 53:101339. [DOI: 10.1016/j.smrv.2020.101339] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/21/2022]
|
36
|
Kayser RR, Haney M, Raskin M, Arout C, Simpson HB. Acute effects of cannabinoids on symptoms of obsessive-compulsive disorder: A human laboratory study. Depress Anxiety 2020; 37:801-811. [PMID: 32383271 PMCID: PMC7423713 DOI: 10.1002/da.23032] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 04/01/2020] [Accepted: 04/19/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Preclinical data implicate the endocannabinoid system in the pathology underlying obsessive-compulsive disorder (OCD), while survey data have linked OCD symptoms to increased cannabis use. Cannabis products are increasingly marketed as treatments for anxiety and other OCD-related symptoms. Yet, few studies have tested the acute effects of cannabis on psychiatric symptoms in humans. METHODS We recruited 14 adults with OCD and prior experience using cannabis to enter a randomized, placebo-controlled, human laboratory study to compare the effects on OCD symptoms of cannabis containing varying concentrations of Δ-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) on OCD symptoms to placebo. We used a within-subjects design to increase statistical power. Across three laboratory sessions, participants smoked three cannabis varietals in random order: placebo (0% THC/0% CBD); THC (7.0% THC/0.18% CBD); and CBD (0.4% THC/10.4% CBD). We analyzed acute changes in OCD symptoms, state anxiety, cardiovascular measures, and drug-related effects (e.g., euphoria) as a function of varietal. RESULTS Twelve participants completed the study. THC increased heart rate, blood pressure, and intoxication compared with CBD and placebo. Self-reported OCD symptoms and anxiety decreased over time in all three conditions. Although OCD symptoms did not vary as a function of cannabis varietal, state anxiety was significantly lower immediately after placebo administration relative to both THC and CBD. CONCLUSIONS This is the first placebo-controlled investigation of cannabis in adults with OCD. The data suggest that smoked cannabis, whether containing primarily THC or CBD, has little acute impact on OCD symptoms and yields smaller reductions in anxiety compared to placebo.
Collapse
Affiliation(s)
- Reilly R. Kayser
- Department of Psychiatry, Columbia University Vagelos
College of Physicians and Surgeons, New York, NY,Research Foundation for Mental Hygiene, New York State
Psychiatric Institute, New York, NY
| | - Margaret Haney
- Department of Psychiatry, Columbia University Vagelos
College of Physicians and Surgeons, New York, NY,Research Foundation for Mental Hygiene, New York State
Psychiatric Institute, New York, NY
| | - Marissa Raskin
- Research Foundation for Mental Hygiene, New York State
Psychiatric Institute, New York, NY
| | - Caroline Arout
- Department of Psychiatry, Columbia University Vagelos
College of Physicians and Surgeons, New York, NY,Research Foundation for Mental Hygiene, New York State
Psychiatric Institute, New York, NY
| | - H. Blair Simpson
- Department of Psychiatry, Columbia University Vagelos
College of Physicians and Surgeons, New York, NY,Research Foundation for Mental Hygiene, New York State
Psychiatric Institute, New York, NY
| |
Collapse
|
37
|
Kesner AJ, Lovinger DM. Cannabinoids, Endocannabinoids and Sleep. Front Mol Neurosci 2020; 13:125. [PMID: 32774241 PMCID: PMC7388834 DOI: 10.3389/fnmol.2020.00125] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/22/2020] [Indexed: 12/21/2022] Open
Abstract
Sleep is a vital function of the nervous system that contributes to brain and bodily homeostasis, energy levels, cognitive ability, and other key functions of a variety of organisms. Dysfunctional sleep induces neural problems and is a key part of almost all human psychiatric disorders including substance abuse disorders. The hypnotic effects of cannabis have long been known and there is increasing use of phytocannabinoids and other formulations as sleep aids. Thus, it is crucial to gain a better understanding of the neurobiological basis of cannabis drug effects on sleep, as well as the role of the endogenous cannabinoid system in sleep physiology. In this review article, we summarize the current state of knowledge concerning sleep-related endogenous cannabinoid function derived from research on humans and rodent models. We also review information on acute and chronic cannabinoid drug effects on sleep in these organisms, and molecular mechanisms that may contribute to these effects. We point out the potential benefits of acute cannabinoids for sleep improvement, but also the potential sleep-disruptive effects of withdrawal following chronic cannabinoid drug use. Prescriptions for future research in this burgeoning field are also provided.
Collapse
Affiliation(s)
- Andrew J Kesner
- Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institute of Health (NIH), Bethesda, MD, United States
- Center on Compulsive Behaviors, Intramural Research Program, National Institute of Health (NIH), Bethesda, MD, United States
| | - David M Lovinger
- Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institute of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
38
|
Perogamvros L, Castelnovo A, Samson D, Dang-Vu TT. Failure of fear extinction in insomnia: An evolutionary perspective. Sleep Med Rev 2020; 51:101277. [DOI: 10.1016/j.smrv.2020.101277] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/07/2020] [Accepted: 01/13/2020] [Indexed: 12/22/2022]
|
39
|
Suraev A, Grunstein RR, Marshall NS, D'Rozario AL, Gordon CJ, Bartlett DJ, Wong K, Yee BJ, Vandrey R, Irwin C, Arnold JC, McGregor IS, Hoyos CM. Cannabidiol (CBD) and Δ 9-tetrahydrocannabinol (THC) for chronic insomnia disorder ('CANSLEEP' trial): protocol for a randomised, placebo-controlled, double-blinded, proof-of-concept trial. BMJ Open 2020; 10:e034421. [PMID: 32430450 PMCID: PMC7239553 DOI: 10.1136/bmjopen-2019-034421] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/06/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Insomnia is a highly prevalent and costly condition that is associated with increased health risks and healthcare utilisation. Anecdotally, cannabis use is frequently reported by consumers to promote sleep. However, there is limited research on the effects of cannabis on sleep and daytime function in people with insomnia disorder using objective measures. This proof-of-concept study will evaluate the effects of a single dose of an oral cannabis-based medicine on sleep and daytime function in participants with chronic insomnia disorder. METHODS AND ANALYSIS A randomised, crossover, placebo-controlled, single-dose study design will be used to test the safety and efficacy of an oral oil solution ('ETC120') containing 10 mg Δ9-tetrahydrocannabinol (THC) and 200 mg cannabidiol (CBD) in 20 participants diagnosed with chronic insomnia disorder. Participants aged 35-60 years will be recruited over an 18-month period commencing August 2019. Each participant will receive both the active drug and matched placebo, in a counterbalanced order, during two overnight study assessment visits, with at least a 1-week washout period between each visit. The primary outcomes are total sleep time and wake after sleep onset assessed via polysomnography. In addition, 256-channel high-density electroencephalography and source modelling using structural brain MRI will be used to comprehensively examine brain activation during sleep and wake periods on ETC120 versus placebo. Next-day cognitive function, alertness and simulated driving performance will also be investigated. ETHICS AND DISSEMINATION Ethics approval was received from Bellberry Human Research Ethics Committee (2018-04-284). The findings will be disseminated in a peer-reviewed open-access journal and at academic conferences. TRIAL REGISTRATION NUMBER ANZCTRN12619000714189.
Collapse
Affiliation(s)
- Anastasia Suraev
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Sydney, New South Wales, Australia
- The University of Sydney, Lambert Initiative for Cannabinoid Therapeutics, Sydney, New South Wales, Australia
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, New South Wales, Australia
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia
| | - Ronald R Grunstein
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Sydney, New South Wales, Australia
- RPA-Charles Perkins Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Nathaniel S Marshall
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Sydney, New South Wales, Australia
- The University of Sydney, Faculty of Medicine and Health, Susan Wakil School of Nursing and Midwifery, Sydney, New South Wales, Australia
| | - Angela L D'Rozario
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Sydney, New South Wales, Australia
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, New South Wales, Australia
| | - Christopher J Gordon
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Sydney, New South Wales, Australia
- The University of Sydney, Faculty of Medicine and Health, Susan Wakil School of Nursing and Midwifery, Sydney, New South Wales, Australia
| | - Delwyn J Bartlett
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Sydney, New South Wales, Australia
| | - Keith Wong
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Sydney, New South Wales, Australia
- RPA-Charles Perkins Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Brendon J Yee
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Sydney, New South Wales, Australia
- RPA-Charles Perkins Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Ryan Vandrey
- Behavioral Pharmacology Research Unit, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chris Irwin
- Menzies Health Institute Queensland, School Allied Health Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Jonathon C Arnold
- The University of Sydney, Lambert Initiative for Cannabinoid Therapeutics, Sydney, New South Wales, Australia
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia
- The University of Sydney, Faculty of Medicine and Health, Discipline of Pharmacology, Sydney, New South Wales, Australia
| | - Iain S McGregor
- The University of Sydney, Lambert Initiative for Cannabinoid Therapeutics, Sydney, New South Wales, Australia
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, New South Wales, Australia
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia
| | - Camilla M Hoyos
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Sydney, New South Wales, Australia
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, New South Wales, Australia
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Zhang J, Zhang L, Chang Y, Gu Q, Zhang J, Zhu Z, Qian Z, Wei C, Liu Z, Ren W, Han J. The Endocannabinoid System Contributes to Memory Deficits Induced by Rapid-eye-movement Sleep Deprivation in Adolescent Mice. Neuroscience 2020; 433:174-183. [PMID: 32198011 DOI: 10.1016/j.neuroscience.2020.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 02/07/2023]
Abstract
Sleep loss or insomnia is among the contributing factors of cognitive deficit, the underlying mechanisms of which remain largely elusive. The endocannabinoid (eCB) system plays a role in sleep, while it is unknown if it is involved in the regulation of memory retrieval by sleep deprivation. In addition, it still controversial how rapid-eye-movement sleep deprivation (REMSD) affects the spatial memory of adolescent mice. Here, we found that 24-h REMSD impairs spatial memory retrieval of adolescent mice in an object-place recognition task, which was rescued by NESS0327, a neutral cannabinoid receptor 1 (CB1R) antagonist. Mechanistically, REMSD induced eCB-mediated short-term and long-term synaptic plasticity changing including depolarization-induced suppression of inhibition (DSI) in the pyramidal neurons of the hippocampus, in which long-term synaptic plasticity changing was rescued by NESS0327. REMSD downregulated monoacylglycerol lipase, a hydrolase for the endocannabinoid 2-arachidonoylglycerol (2-AG), suggesting the involvement of eCB accumulation and the consequent synaptic plasticity in REMSD-elicited memory impairment in adolescent mice. These findings shed light on the role of sleep disorders in learning and memory deficit of adolescents.
Collapse
Affiliation(s)
- Jinming Zhang
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Lizi Zhang
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China; College of Life Sciences, Shaanxi Normal University, China
| | - Yuan Chang
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Qiaofen Gu
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Junmin Zhang
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Zhou Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Zhaoqiang Qian
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Chunling Wei
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Zhiqiang Liu
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Wei Ren
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Jing Han
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China.
| |
Collapse
|
41
|
Wyatt RM, Fraser I, Welty N, Lord B, Wennerholm M, Sutton S, Ameriks MK, Dugovic C, Yun S, White A, Nguyen L, Koudriakova T, Tian G, Suarez J, Szewczuk L, Bonnette W, Ahn K, Ghosh B, Flores CM, Connolly PJ, Zhu B, Macielag MJ, Brandt MR, Chevalier K, Zhang SP, Lovenberg T, Bonaventure P. Pharmacologic Characterization of JNJ-42226314, [1-(4-Fluorophenyl)indol-5-yl]-[3-[4-(thiazole-2-carbonyl)piperazin-1-yl]azetidin-1-yl]methanone, a Reversible, Selective, and Potent Monoacylglycerol Lipase Inhibitor. J Pharmacol Exp Ther 2020; 372:339-353. [PMID: 31818916 DOI: 10.1124/jpet.119.262139] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/01/2019] [Indexed: 12/14/2022] Open
Abstract
The serine hydrolase monoacylglycerol lipase (MAGL) is the rate-limiting enzyme responsible for the degradation of the endocannabinoid 2-arachidonoylglycerol (2-AG) into arachidonic acid and glycerol. Inhibition of 2-AG degradation leads to elevation of 2-AG, the most abundant endogenous agonist of the cannabinoid receptors (CBs) CB1 and CB2. Activation of these receptors has demonstrated beneficial effects on mood, appetite, pain, and inflammation. Therefore, MAGL inhibitors have the potential to produce therapeutic effects in a vast array of complex human diseases. The present report describes the pharmacologic characterization of [1-(4-fluorophenyl)indol-5-yl]-[3-[4-(thiazole-2-carbonyl)piperazin-1-yl]azetidin-1-yl]methanone (JNJ-42226314), a reversible and highly selective MAGL inhibitor. JNJ-42226314 inhibits MAGL in a competitive mode with respect to the 2-AG substrate. In rodent brain, the compound time- and dose-dependently bound to MAGL, indirectly led to CB1 occupancy by raising 2-AG levels, and raised norepinephrine levels in cortex. In vivo, the compound exhibited antinociceptive efficacy in both the rat complete Freund's adjuvant-induced radiant heat hypersensitivity and chronic constriction injury-induced cold hypersensitivity models of inflammatory and neuropathic pain, respectively. Though 30 mg/kg induced hippocampal synaptic depression, altered sleep onset, and decreased electroencephalogram gamma power, 3 mg/kg still provided approximately 80% enzyme occupancy, significantly increased 2-AG and norepinephrine levels, and produced neuropathic antinociception without synaptic depression or decreased gamma power. Thus, it is anticipated that the profile exhibited by this compound will allow for precise modulation of 2-AG levels in vivo, supporting potential therapeutic application in several central nervous system disorders. SIGNIFICANCE STATEMENT: Potentiation of endocannabinoid signaling activity via inhibition of the serine hydrolase monoacylglycerol lipase (MAGL) is an appealing strategy in the development of treatments for several disorders, including ones related to mood, pain, and inflammation. [1-(4-Fluorophenyl)indol-5-yl]-[3-[4-(thiazole-2-carbonyl)piperazin-1-yl]azetidin-1-yl]methanone is presented in this report to be a novel, potent, selective, and reversible noncovalent MAGL inhibitor that demonstrates dose-dependent enhancement of the major endocannabinoid 2-arachidonoylglycerol as well as efficacy in models of neuropathic and inflammatory pain.
Collapse
MESH Headings
- Animals
- Binding, Competitive
- Brain/drug effects
- Brain/enzymology
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/blood
- Enzyme Inhibitors/pharmacology
- Escherichia coli/enzymology
- Escherichia coli/genetics
- HeLa Cells
- Humans
- Kinetics
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/enzymology
- Male
- Mice, Inbred C57BL
- Molecular Structure
- Monoacylglycerol Lipases/antagonists & inhibitors
- Monoacylglycerol Lipases/genetics
- Pain/drug therapy
- Piperazines/blood
- Piperazines/pharmacology
- Protein Binding
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB2/agonists
- Sleep, REM/drug effects
- Substrate Specificity
Collapse
Affiliation(s)
- Ryan M Wyatt
- Janssen Research & Development, LLC, San Diego, California
| | - Ian Fraser
- Janssen Research & Development, LLC, San Diego, California
| | - Natalie Welty
- Janssen Research & Development, LLC, San Diego, California
| | - Brian Lord
- Janssen Research & Development, LLC, San Diego, California
| | | | - Steven Sutton
- Janssen Research & Development, LLC, San Diego, California
| | | | | | - Sujin Yun
- Janssen Research & Development, LLC, San Diego, California
| | - Allison White
- Janssen Research & Development, LLC, San Diego, California
| | - Leslie Nguyen
- Janssen Research & Development, LLC, San Diego, California
| | | | - Gaochao Tian
- Janssen Research & Development, LLC, San Diego, California
| | - Javier Suarez
- Janssen Research & Development, LLC, San Diego, California
| | | | | | - Kay Ahn
- Janssen Research & Development, LLC, San Diego, California
| | - Brahma Ghosh
- Janssen Research & Development, LLC, San Diego, California
| | | | | | - Bin Zhu
- Janssen Research & Development, LLC, San Diego, California
| | | | | | | | - Sui-Po Zhang
- Janssen Research & Development, LLC, San Diego, California
| | | | | |
Collapse
|
42
|
Antony JM, McDonald AC, Noorbakhsh F, Guthrie N, Evans M. Changing the Tone of Clinical Study Design in the Cannabis Industry. Transl Neurosci 2020; 11:4-9. [PMID: 32104589 PMCID: PMC7029654 DOI: 10.1515/tnsci-2020-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/16/2020] [Indexed: 12/17/2022] Open
Abstract
Cannabis (also known as marijuana) is the most frequently used psychoactive substance globally. Cannabis exerts therapeutic functions for many indications and has vast potential as a health and wellness product. Advances in our understanding of the composition and pharmacological properties of cannabis have revealed interactions between cannabis, an individuals’ circadian rhythms and their endocannabinoid signaling. Exogenously administered cannabinoids can bidirectionally entrain central and peripheral clocks that comprise circadian rhythms, and malfunctions in the endocannabinoid system are reported to impact neurological processes. Therefore, it is necessary to account for the circadian rhythm when designing clinical trials examining the pharmacological properties of cannabis-based products for health and wellness to limit its potential confounding impact on results. Consideration of the entrainment capabilities of the endocannabinoid system is warranted when designing clinical trials.
Collapse
Affiliation(s)
| | | | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
43
|
Abrahao KP, Pava MJ, Lovinger DM. Dose-dependent alcohol effects on electroencephalogram: Sedation/anesthesia is qualitatively distinct from sleep. Neuropharmacology 2019; 164:107913. [PMID: 31843396 DOI: 10.1016/j.neuropharm.2019.107913] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/27/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023]
Abstract
Alcohol is commonly used as a sleep inducer/aid by humans. However, individuals diagnosed with alcohol use disorders have sleep problems. Few studies have examined the effect of ethanol on physiological features of sedation and anesthesia, particularly at high doses. This study used polysomnography and a rapid, unbiased scoring of vigilance states with an automated algorithm to provide a thorough characterization of dose-dependent acute ethanol effects on sleep and electroencephalogram (EEG) power spectra in C57BL/6J male mice. Ethanol had a narrow dose-response effect on sleep. Only a high dose (4.0 g/kg) produced a unique, transient state that could not be characterized in terms of canonical sleep-wake states, so we dubbed this novel state Drug-Induced State with a Characteristic Oscillation in the Theta Band (DISCO-T). After this anesthetic effect, the high dose of alcohol promoted NREM sleep by increasing the duration of NREM bouts while reducing wake. REM sleep was differentially responsive to the circadian timing of ethanol administration. EEG power spectra proved more sensitive to ethanol than sleep measures as there were clear effects of ethanol at 2.0 and 4.0 g/kg doses. Ethanol promoted delta oscillations and suppressed faster frequencies, but there were clear, differential effects on wake and REM EEG power based on the timing of the ethanol injection. Understanding the neural basis of the extreme soporific effects of high dose ethanol may aid in treating acute toxicity brought about by patterns of excessive binge consumption commonly observed in young people.
Collapse
Affiliation(s)
- Karina P Abrahao
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| | - Matthew J Pava
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
44
|
González J, Cavelli M, Mondino A, Pascovich C, Castro-Zaballa S, Torterolo P, Rubido N. Decreased electrocortical temporal complexity distinguishes sleep from wakefulness. Sci Rep 2019; 9:18457. [PMID: 31804569 PMCID: PMC6895088 DOI: 10.1038/s41598-019-54788-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/06/2019] [Indexed: 11/09/2022] Open
Abstract
In most mammals, the sleep-wake cycle is constituted by three behavioral states: wakefulness (W), non-REM (NREM) sleep, and REM sleep. These states are associated with drastic changes in cognitive capacities, mostly determined by the function of the thalamo-cortical system. The intra-cranial electroencephalogram or electocorticogram (ECoG), is an important tool for measuring the changes in the thalamo-cortical activity during W and sleep. In the present study we analyzed broad-band ECoG recordings of the rat by means of a time-series complexity measure that is easy to implement and robust to noise: the Permutation Entropy (PeEn). We found that PeEn is maximal during W and decreases during sleep. These results bring to light the different thalamo-cortical dynamics emerging during sleep-wake states, which are associated with the well-known spectral changes that occur when passing from W to sleep. Moreover, the PeEn analysis allows us to determine behavioral states independently of the electrodes' cortical location, which points to an underlying global pattern in the signal that differs among the cycle states that is missed by classical methods. Consequently, our data suggest that PeEn analysis of a single EEG channel could allow for cheap, easy, and efficient sleep monitoring.
Collapse
Affiliation(s)
- Joaquín González
- Universidad de la República, Departamento de Fisiología de Facultad de Medicina, Av. Gral. Flores 2125, 11800, Montevideo, Uruguay
| | - Matias Cavelli
- Universidad de la República, Departamento de Fisiología de Facultad de Medicina, Av. Gral. Flores 2125, 11800, Montevideo, Uruguay
| | - Alejandra Mondino
- Universidad de la República, Departamento de Fisiología de Facultad de Medicina, Av. Gral. Flores 2125, 11800, Montevideo, Uruguay
| | - Claudia Pascovich
- Universidad de la República, Departamento de Fisiología de Facultad de Medicina, Av. Gral. Flores 2125, 11800, Montevideo, Uruguay
| | - Santiago Castro-Zaballa
- Universidad de la República, Departamento de Fisiología de Facultad de Medicina, Av. Gral. Flores 2125, 11800, Montevideo, Uruguay
| | - Pablo Torterolo
- Universidad de la República, Departamento de Fisiología de Facultad de Medicina, Av. Gral. Flores 2125, 11800, Montevideo, Uruguay.
| | - Nicolás Rubido
- Universidad de la República, Instituto de Física de Facultad de Ciencias, Iguá 4225, 11400, Montevideo, Uruguay
| |
Collapse
|
45
|
Santori A, Colucci P, Mancini GF, Morena M, Palmery M, Trezza V, Puglisi-Allegra S, Hill MN, Campolongo P. Anandamide modulation of circadian- and stress-dependent effects on rat short-term memory. Psychoneuroendocrinology 2019; 108:155-162. [PMID: 31302498 DOI: 10.1016/j.psyneuen.2019.06.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 05/22/2019] [Accepted: 06/28/2019] [Indexed: 10/26/2022]
Abstract
The endocannabinoid system plays a key role in the control of emotional responses to environmental challenges. CB1 receptors are highly expressed within cortico-limbic brain areas, where they modulate stress effects on memory processes. Glucocorticoid and endocannabinoid release is influenced by circadian rhythm. Here, we investigated how different stress intensities immediately after encoding influence rat short-term memory in an object recognition task, whether the effects depend on circadian rhythm and if exogenous augmentation of anandamide levels could restore any observed impairment. Two separate cohorts of male adult Sprague-Dawley rats were tested at two different times of the day, morning (inactivity phase) or afternoon (before the onset of the activity phase) in an object recognition task. The anandamide hydrolysis inhibitor URB597 was intraperitoneally administered immediately after the training trial. Rats were thereafter subjected to a forced swim stress under low or high stress conditions and tested 1 h after training. Control rats underwent the same experimental procedure except for the forced swim stress (no stress). We further investigated whether URB597 administration might modulate corticosterone release in rats subjected to the different stress conditions, both in the morning or afternoon. The low stressor elevated plasma corticosterone levels and impaired 1 h recognition memory performance when animals were tested in the morning. Exposure to the higher stress condition elevated plasma corticosterone levels and impaired memory performance, independently of the testing time. These findings show that stress impairing effects on short-term recognition memory are dependent on the intensity of stress and circadian rhythm. URB597 (0.3 mg kg-1) rescued the altered memory performance and decreased corticosterone levels in all the impaired groups yet leaving memory unaltered in the non-impaired groups.
Collapse
Affiliation(s)
- Alessia Santori
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy
| | - Paola Colucci
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy
| | | | - Maria Morena
- Hotchkiss Brain Institute, Depts. of Cell Biology and Anatomy &Psychiatry, University of Calgary, T2N 4N1, Calgary, Canada
| | - Maura Palmery
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy
| | - Viviana Trezza
- Dept. of Science, Section of Biomedical Sciences and Technologies, University Roma Tre, 00146, Rome, Italy
| | | | - Matthew N Hill
- Hotchkiss Brain Institute, Depts. of Cell Biology and Anatomy &Psychiatry, University of Calgary, T2N 4N1, Calgary, Canada
| | - Patrizia Campolongo
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy; Neurobiology of Behavior Laboratory, Santa Lucia Foundation, 00143, Rome, Italy.
| |
Collapse
|
46
|
Li X, Vigil JM, Stith SS, Brockelman F, Keeling K, Hall B. The effectiveness of self-directed medical cannabis treatment for pain. Complement Ther Med 2019; 46:123-130. [DOI: 10.1016/j.ctim.2019.07.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/16/2019] [Accepted: 07/26/2019] [Indexed: 12/23/2022] Open
|
47
|
Minichino A, Senior M, Brondino N, Zhang SH, Godwlewska BR, Burnet PW, Cipriani A, Lennox BR. Measuring Disturbance of the Endocannabinoid System in Psychosis: A Systematic Review and Meta-analysis. JAMA Psychiatry 2019; 76:914-923. [PMID: 31166595 PMCID: PMC6552109 DOI: 10.1001/jamapsychiatry.2019.0970] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
IMPORTANCE The endocannabinoid system (ECS) is a lipid-based endogenous signaling system. Its relevance to psychosis is through the association between cannabis use and the onset and course of illness and through the antipsychotic properties of cannabidiol, a potential ECS enhancer. OBJECTIVE To conduct a systematic review and meta-analysis of the blood and cerebrospinal fluid (CSF) measures of the ECS in psychotic disorders. DATA SOURCES Web of Science and PubMed were searched from inception through June 13, 2018. The articles identified were reviewed, as were citations to previous publications and the reference lists of retrieved articles. STUDY SELECTION Original articles were included that reported blood or CSF measures of ECS activity in patients with psychotic illnesses and in healthy controls. DATA EXTRACTION AND SYNTHESIS PRISMA guidelines, independent extraction by multiple observers, and random-effects meta-analysis were used. Heterogeneity was assessed with the I2 index. Sensitivity analyses tested the robustness of the results. MAIN OUTCOMES AND MEASURES The clinical relevance of ECS modifications in psychotic disorders was investigated by (1) a quantitative synthesis of the differences in blood and CSF markers of the ECS between patients and healthy controls, and (2) a qualitative synthesis of the association of these markers with symptom severity, stage of illness, and response to treatment. RESULTS A total of 18 studies were included. Three individual meta-analyses were performed to identify the differences in ECS markers between people with schizophrenia and healthy controls. Five studies, including 226 patients and 385 controls, reported significantly higher concentrations of anandamide in the CSF of patients than controls (standardized mean difference [SMD], 0.97; 95% CI, 0.67-1.26; P < .001; I2 = 54.8%). In 9 studies, with 344 patients and 411 controls, significantly higher anandamide levels in blood were found in patients, compared with controls (SMD, 0.55; 95% CI, 0.05-1.04; P = .03; I2 = 89.6%). In 3 studies, involving 88 patients and 179 controls, a significantly higher expression of type 1 cannabinoid receptors on peripheral immune cells was reported in patients compared with controls (SMD, 0.57; 95% CI, 0.31-0.84; P < .001; I2 = 0%). Higher ECS tone was found at an early stage of illness in individuals who were antipsychotic naïve or free, and it had an inverse association with symptom severity and was normalized after successful treatment. Moderate to high level of heterogeneity in methods was found between studies. CONCLUSIONS AND RELEVANCE Testing clinically relevant markers of the ECS in the blood and CSF of people with psychotic illness appears possible, and these markers provide useful biomarkers for the psychotic disorder; however, not all studies accounted for important variables, such as cannabis use. TRIAL REGISTRATION PROSPERO identifier: CRD42018099863.
Collapse
Affiliation(s)
- Amedeo Minichino
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Morwenna Senior
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Natascia Brondino
- Section of Psychiatry, Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Sam H Zhang
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | | | - Philip W.J Burnet
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Andrea Cipriani
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom,Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, United Kingdom
| | - Belinda R. Lennox
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
48
|
Peball M, Werkmann M, Ellmerer P, Stolz R, Valent D, Knaus HG, Ulmer H, Djamshidian A, Poewe W, Seppi K. Nabilone for non-motor symptoms of Parkinson's disease: a randomized placebo-controlled, double-blind, parallel-group, enriched enrolment randomized withdrawal study (The NMS-Nab Study). J Neural Transm (Vienna) 2019; 126:1061-1072. [PMID: 31129719 PMCID: PMC6647387 DOI: 10.1007/s00702-019-02021-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 05/22/2019] [Indexed: 01/07/2023]
Abstract
Although open-label observations report a positive effect of cannabinoids on non-motor symptoms (NMS) in Parkinson's disease (PD) patients, these effects remain to be investigated in a controlled trial for a broader use in NMS in PD patients. Therefore, we decided to design a proof-of-concept study to assess the synthetic cannabinoid nabilone for the treatment of NMS. We hypothesize that nabilone will improve NMS in patients with PD and have a favorable safety profile. The NMS-Nab Study is as a mono-centric phase II, randomized, placebo-controlled, double-blind, parallel-group, enriched enrollment withdrawal study. The primary efficacy criterion will be the change in Movement Disorders Society-Unified Parkinson's Disease-Rating Scale Part I score between baseline (i.e. randomization) and week 4. A total of 38 patients will have 80% power to detect a probability of 0.231 that an observation in the treatment group is less than an observation in the placebo group using a Wilcoxon rank-sum test with a 0.050 two-sided significance level assuming a true difference of 2.5 points between nabilone and placebo in the primary outcome measure and a standard deviation of the change of 2.4 points. The reduction of harm through an ineffective treatment, the possibility of individualized dosing, the reduction of sample size, and the possible evaluation of the influence of the placebo effect on efficacy outcomes justify this design for a single-centered placebo-controlled investigator-initiated trial of nabilone. This study should be the basis for further evaluations of long-term efficacy and safety of the use of cannabinoids in PD patients.
Collapse
Affiliation(s)
- Marina Peball
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Mario Werkmann
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Philipp Ellmerer
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Raphaela Stolz
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Dora Valent
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Hans-Günther Knaus
- Department for Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, Peter-Mayr Straße 1, 6020, Innsbruck, Austria
| | - Hanno Ulmer
- Department of Medical Statistics, Informatics and Health Economics, Innsbruck Medical University, Schöpfstraße 41/1, 6020, Innsbruck, Austria
| | - Atbin Djamshidian
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Werner Poewe
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Klaus Seppi
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
49
|
Martin SC, Monroe SK, Diering GH. Homer1a and mGluR1/5 Signaling in Homeostatic Sleep Drive and Output. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2019; 92:93-101. [PMID: 30923476 PMCID: PMC6430175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Sleep is an essential physiological behavior that promotes cognitive development and function. Although the switch between sleep/wake cycles is controlled by specific neural circuits, sleep need and the restorative benefits of sleep are likely controlled by cellular mechanisms localized in critical areas of the brain involved in learning and memory including the cortex and hippocampus. However, the molecular basis for the restorative function(s) of sleep that support cognition, or for the homeostatic build-up of sleep need are poorly understood. Synapses undergo local and global changes in strength to support learning and memory and are likely a point of restoration during sleep. Homer1a and mGluR1/5, recently implicated in sleep function, are molecules involved in the scaling down process that weakens synapses during sleep to restore synapse homeostasis. During wake, long-form Homer proteins tether mGluR1/5 to IP3R and to the post-synaptic density (PSD). During sleep, short-form Homer1a uncouples mGluR1/5 from IP3R leaving mGluR1/5 open to interact with other effectors, switching mGluR1/5 signaling from "awake-type" to "sleep-type" signaling modes. Importantly, mGluR1/5 have been implicated in several neurological and neurodevelopmental disorders such as Alzheimer's disease (AD) and autism spectrum disorder (ASD), all of which show abnormal sleep phenotypes, linking sleep, disease, and mGluR1/5 signaling. Further investigation into the downstream effectors of mGluR1/5 and sleep/wake signaling will lead to more targeted therapeutic interventions.
Collapse
Affiliation(s)
- Shenée C. Martin
- Neuroscience Center and Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, NC
| | - Sarah K. Monroe
- Neuroscience Center and Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, NC
| | - Graham H. Diering
- Neuroscience Center and Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, NC,Carolina Institute for Developmental Disabilities, University of North Carolina Chapel Hill, NC,To whom all correspondence should be addressed: Graham H. Diering, University of North Carolina 111 Mason Farm Road, 5200 Medical Biomolecular Research Building, Chapel Hill, NC 27599-7545; Tel: 919-966-1464, Fax: 919-966-6927,
| |
Collapse
|
50
|
Stith SS, Vigil JM, Brockelman F, Keeling K, Hall B. Patient-Reported Symptom Relief Following Medical Cannabis Consumption. Front Pharmacol 2018; 9:916. [PMID: 30210337 PMCID: PMC6121171 DOI: 10.3389/fphar.2018.00916] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/26/2018] [Indexed: 01/04/2023] Open
Abstract
Background: The Releaf AppTM mobile software application (app) data was used to measure self-reported effectiveness and side effects of medical cannabis used under naturalistic conditions. Methods: Between 5/03/2016 and 12/16/2017, 2,830 Releaf AppTM users completed 13,638 individual sessions self-administering medical cannabis and indicated their primary health symptom severity rating on an 11-point (0–10) visual analog scale in real-time prior to and following cannabis consumption, along with experienced side effects. Results: Releaf AppTM responders used cannabis to treat myriad health symptoms, the most frequent relating to pain, anxiety, and depressive conditions. Significant symptom severity reductions were reported for all the symptom categories, with mean reductions between 2.8 and 4.6 points (ds ranged from 1.29–2.39, ps < 0.001). On average, higher pre-dosing symptom levels were associated with greater reported symptom relief, and users treating anxiety or depression-related symptoms reported significantly more relief (ps < 0.001) than users with pain symptoms. Of the 42 possible side effects, users were more likely to indicate and showed a stronger correlation between symptom relief and experiences of positive (94% of sessions) or a context-specific side effects (76%), whereas negative side effects (60%) were associated with lessened, yet still significant symptom relief and were more common among patients treating a depressive symptom relative to patients treating anxiety and pain-related conditions. Conclusion: Patient-managed cannabis use is associated with clinically significant improvements in self-reported symptom relief for treating a wide range of health conditions, along with frequent positive and negative side effects.
Collapse
Affiliation(s)
- Sarah S Stith
- Department of Economics, The University of New Mexico, Albuquerque, NM, United States
| | - Jacob M Vigil
- Department of Psychology, The University of New Mexico, Albuquerque, NM, United States
| | | | | | - Branden Hall
- The MoreBetter Ltd., Washington, DC, United States
| |
Collapse
|