1
|
Sharma P, Nelson RJ. Disrupted Circadian Rhythms and Substance Use Disorders: A Narrative Review. Clocks Sleep 2024; 6:446-467. [PMID: 39189197 PMCID: PMC11348162 DOI: 10.3390/clockssleep6030030] [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: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/28/2024] Open
Abstract
Substance use disorder is a major global health concern, with a high prevalence among adolescents and young adults. The most common substances of abuse include alcohol, marijuana, cocaine, nicotine, and opiates. Evidence suggests that a mismatch between contemporary lifestyle and environmental demands leads to disrupted circadian rhythms that impair optimal physiological and behavioral function, which can increase the vulnerability to develop substance use disorder and related problems. The circadian system plays an important role in regulating the sleep-wake cycle and reward processing, both of which directly affect substance abuse. Distorted substance use can have a reciprocal effect on the circadian system by influencing circadian clock gene expression. Considering the detrimental health consequences and profound societal impact of substance use disorder, it is crucial to comprehend its complex association with circadian rhythms, which can pave the way for the generation of novel chronotherapeutic treatment approaches. In this narrative review, we have explored the potential contributions of disrupted circadian rhythms and sleep on use and relapse of different substances of abuse. The involvement of circadian clock genes with drug reward pathways is discussed, along with the potential research areas that can be explored to minimize disordered substance use by improving circadian hygiene.
Collapse
Affiliation(s)
- Pallavi Sharma
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA;
| | | |
Collapse
|
2
|
Camberos-Barraza J, Camacho-Zamora A, Bátiz-Beltrán JC, Osuna-Ramos JF, Rábago-Monzón ÁR, Valdez-Flores MA, Angulo-Rojo CE, Guadrón-Llanos AM, Picos-Cárdenas VJ, Calderón-Zamora L, Norzagaray-Valenzuela CD, Cárdenas-Torres FI, De la Herrán-Arita AK. Sleep, Glial Function, and the Endocannabinoid System: Implications for Neuroinflammation and Sleep Disorders. Int J Mol Sci 2024; 25:3160. [PMID: 38542134 PMCID: PMC10970053 DOI: 10.3390/ijms25063160] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 11/11/2024] Open
Abstract
The relationship between sleep, glial cells, and the endocannabinoid system represents a multifaceted regulatory network with profound implications for neuroinflammation and cognitive function. The molecular underpinnings of sleep modulation by the endocannabinoid system and its influence on glial cell activity are discussed, shedding light on the reciprocal relationships that govern these processes. Emphasis is placed on understanding the role of glial cells in mediating neuroinflammatory responses and their modulation by sleep patterns. Additionally, this review examines how the endocannabinoid system interfaces with glia-immune signaling to regulate inflammatory cascades within the central nervous system. Notably, the cognitive consequences of disrupted sleep, neuroinflammation, and glial dysfunction are addressed, encompassing implications for neurodegenerative disorders, mood disturbances, and cognitive decline. Insights into the bidirectional modulation of cognitive function by the endocannabinoid system in the context of sleep and glial activity are explored, providing a comprehensive perspective on the potential mechanisms underlying cognitive impairments associated with sleep disturbances. Furthermore, this review examines potential therapeutic avenues targeting the endocannabinoid system to mitigate neuroinflammation, restore glial homeostasis, and normalize sleep patterns. The identification of novel therapeutic targets within this intricate regulatory network holds promise for addressing conditions characterized by disrupted sleep, neuroinflammation, and cognitive dysfunction. This work aims to examine the complexities of neural regulation and identify potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
| | | | | | - Juan F. Osuna-Ramos
- Faculty of Medicine, Autonomous University of Sinaloa, Culiacán 80019, Mexico
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
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
|
4
|
Tuvel AL, Winiger EA, Ross JM. A Review of the Effects of Adolescent Cannabis Use on Physical Health. Psychiatr Clin North Am 2023; 46:719-739. [PMID: 37879834 DOI: 10.1016/j.psc.2023.03.005] [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] [Indexed: 10/27/2023]
Abstract
The current review highlights the available research related to cannabis and indicators of physical health in a variety of domains. Various studies have found associations between cannabis use with pulmonary, cardiovascular, gastrointestinal, and endocrine function as well as body mass index and sleep. At this time, more research is needed to understand the influence of cannabis use on physical health, particularly among adolescent samples.
Collapse
Affiliation(s)
- Abigail L Tuvel
- Department of Psychology and Neuroscience, University of Colorado Boulder, 1777 Exposition Drive, Boulder, CO 80301
| | - Evan A Winiger
- Department of Psychiatry, School of Medicine, University of Colorado Anschutz Medical Campus, 1890 N Revere Court, Aurora, CO, 80045
| | - J Megan Ross
- Department of Psychiatry, Division of Addiction Sciences, Treatment and Prevention, University of Colorado Anschutz Medical Campus, 1890 N Revere Court, Aurora, CO, 80045.
| |
Collapse
|
5
|
Salinsky LM, Merritt CR, Zamora JC, Giacomini JL, Anastasio NC, Cunningham KA. μ-opioid receptor agonists and psychedelics: pharmacological opportunities and challenges. Front Pharmacol 2023; 14:1239159. [PMID: 37886127 PMCID: PMC10598667 DOI: 10.3389/fphar.2023.1239159] [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: 06/12/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
Opioid misuse and opioid-involved overdose deaths are a massive public health problem involving the intertwined misuse of prescription opioids for pain management with the emergence of extremely potent fentanyl derivatives, sold as standalone products or adulterants in counterfeit prescription opioids or heroin. The incidence of repeated opioid overdose events indicates a problematic use pattern consistent with the development of the medical condition of opioid use disorder (OUD). Prescription and illicit opioids reduce pain perception by activating µ-opioid receptors (MOR) localized to the central nervous system (CNS). Dysregulation of meso-corticolimbic circuitry that subserves reward and adaptive behaviors is fundamentally involved in the progressive behavioral changes that promote and are consequent to OUD. Although opioid-induced analgesia and the rewarding effects of abused opioids are primarily mediated through MOR activation, serotonin (5-HT) is an important contributor to the pharmacology of opioid abused drugs (including heroin and prescription opioids) and OUD. There is a recent resurgence of interest into psychedelic compounds that act primarily through the 5-HT2A receptor (5-HT 2A R) as a new frontier in combatting such diseases (e.g., depression, anxiety, and substance use disorders). Emerging data suggest that the MOR and 5-HT2AR crosstalk at the cellular level and within key nodes of OUD circuitry, highlighting a major opportunity for novel pharmacological intervention for OUD. There is an important gap in the preclinical profiling of psychedelic 5-HT2AR agonists in OUD models. Further, as these molecules carry risks, additional analyses of the profiles of non-hallucinogenic 5-HT2AR agonists and/or 5-HT2AR positive allosteric modulators may provide a new pathway for 5-HT2AR therapeutics. In this review, we discuss the opportunities and challenges associated with utilizing 5-HT2AR agonists as therapeutics for OUD.
Collapse
Affiliation(s)
| | | | | | | | - Noelle C. Anastasio
- Center for Addiction Sciences and Therapeutics and Department of Pharmacology and Toxicology, John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Kathryn A. Cunningham
- Center for Addiction Sciences and Therapeutics and Department of Pharmacology and Toxicology, John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
6
|
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
|
7
|
Chen J, Garcia EJ, Merritt CR, Zamora JC, Bolinger AA, Pazdrak K, Stafford SJ, Mifflin RC, Wold EA, Wild CT, Chen H, Anastasio NC, Cunningham KA, Zhou J. Discovery of Novel Oleamide Analogues as Brain-Penetrant Positive Allosteric Serotonin 5-HT 2C Receptor and Dual 5-HT 2C/5-HT 2A Receptor Modulators. J Med Chem 2023; 66:9992-10009. [PMID: 37462530 PMCID: PMC10853020 DOI: 10.1021/acs.jmedchem.3c00908] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
The serotonin 5-HT2A receptor (5-HT2AR) and 5-HT2CR localize to the brain and share overlapping signal transduction facets that contribute to their roles in cognition, mood, learning, and memory. Achieving selective targeting of these receptors is challenged by the similarity in their 5-HT orthosteric binding pockets. A fragment-based discovery approach was employed to design and synthesize novel oleamide analogues as selective 5-HT2CR or dual 5-HT2CR/5-HT2AR positive allosteric modulators (PAMs). Compound 13 (JPC0323) exhibited on-target properties, acceptable plasma exposure and brain penetration, as well as negligible displacement to orthosteric sites of ∼50 GPCRs and transporters. Furthermore, compound 13 suppressed novelty-induced locomotor activity in a 5-HT2CR-dependent manner, suggesting 5-HT2CR PAM, but not 5-HT2AR, activity at the level of the whole organism at the employed doses of 13. We discovered new selective 5-HT2CR PAMs and first-in-class 5-HT2CR/5-HT2AR dual PAMs that broaden the pharmacological toolbox to explore the biology of these vital receptors.
Collapse
Affiliation(s)
- Jianping Chen
- Center for Addiction Sciences and Therapeutics and Chemical Biology Program and Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Erik J. Garcia
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Christina R. Merritt
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Joshua C. Zamora
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Andrew A. Bolinger
- Center for Addiction Sciences and Therapeutics and Chemical Biology Program and Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Konrad Pazdrak
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Susan J. Stafford
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Randy C. Mifflin
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Eric A. Wold
- Center for Addiction Sciences and Therapeutics and Chemical Biology Program and Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Christopher T. Wild
- Center for Addiction Sciences and Therapeutics and Chemical Biology Program and Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Haiying Chen
- Center for Addiction Sciences and Therapeutics and Chemical Biology Program and Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Noelle C. Anastasio
- Center for Addiction Sciences and Therapeutics and Chemical Biology Program and Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Kathryn A. Cunningham
- Center for Addiction Sciences and Therapeutics and Chemical Biology Program and Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Jia Zhou
- Center for Addiction Sciences and Therapeutics and Chemical Biology Program and Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| |
Collapse
|
8
|
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
|
9
|
Ray S, Sil S, Kannan M, Periyasamy P, Buch S. Role of the gut-brain axis in HIV and drug abuse-mediated neuroinflammation. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2023; 3:11092. [PMID: 38389809 PMCID: PMC10880759 DOI: 10.3389/adar.2023.11092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/23/2023] [Indexed: 02/24/2024]
Abstract
Drug abuse and related disorders are a global public health crisis affecting millions, but to date, limited treatment options are available. Abused drugs include but are not limited to opioids, cocaine, nicotine, methamphetamine, and alcohol. Drug abuse and human immunodeficiency virus-1/acquired immune deficiency syndrome (HIV-1/AIDS) are inextricably linked. Extensive research has been done to understand the effect of prolonged drug use on neuronal signaling networks and gut microbiota. Recently, there has been rising interest in exploring the interactions between the central nervous system and the gut microbiome. This review summarizes the existing research that points toward the potential role of the gut microbiome in the pathogenesis of HIV-1-linked drug abuse and subsequent neuroinflammation and neurodegenerative disorders. Preclinical data about gut dysbiosis as a consequence of drug abuse in the context of HIV-1 has been discussed in detail, along with its implications in various neurodegenerative disorders. Understanding this interplay will help elucidate the etiology and progression of drug abuse-induced neurodegenerative disorders. This will consequently be beneficial in developing possible interventions and therapeutic options for these drug abuse-related disorders.
Collapse
Affiliation(s)
- Sudipta Ray
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Muthukumar Kannan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
10
|
Tervo-Clemmens B, Schmitt W, Wheeler G, Cooke ME, Schuster RM, Hickey S, Pachas GN, Evins AE, Gilman JM. Cannabis use and sleep quality in daily life: An electronic daily diary study of adults starting cannabis for health concerns. Drug Alcohol Depend 2023; 243:109760. [PMID: 36638745 PMCID: PMC10015315 DOI: 10.1016/j.drugalcdep.2022.109760] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/07/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Real world patterns of cannabis use for health concerns are highly variable and rarely overseen by a physician. Pragmatic effectiveness studies with electronic daily diaries that capture person-specific patterns of cannabis use and health symptoms may help clarify risks and benefits. METHODS As part of a larger, randomized trial (NCT03224468), adults (N = 181) seeking cannabis for insomnia, pain, or anxiety or depressive symptoms were randomized to obtain a medical cannabis card immediately (MCC) or a waitlist control (WLC) and completed 12-weeks of daily web-based surveys on cannabis use and sleep, pain, and depressive symptoms. RESULTS Completion rates of daily surveys were moderate to high (median completed: 72 out of 90 days). Daily reports of cannabis use were consistent with monthly interview assessments and urinalysis. The MCC group increased cannabis use frequency in the 12 weeks following randomization, while WLC did not. Among the MCC group, self-reported sleep quality was significantly higher on cannabis use days, compared to nonuse days. The MCC group displayed long-term sleep improvements, consistent with increasing cannabis frequency. No improvements were found for pain or depressive symptoms. CONCLUSION Cannabis use is associated with same day improvements in self-reported sleep quality, but not pain or depressive symptoms, although sleep improvements occurred in the context of increased frequency of cannabis use, raising the risk for cannabis use disorder. Daily web-based assessments of cannabis appear valid and feasible in adults seeking cannabis for health concerns, providing a flexible, complementary method for future real-world effectiveness studies with expanded and objective measures.
Collapse
Affiliation(s)
- Brenden Tervo-Clemmens
- Center for Addiction Medicine, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - William Schmitt
- Center for Addiction Medicine, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Grace Wheeler
- Center for Addiction Medicine, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Megan E Cooke
- Center for Addiction Medicine, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Randi M Schuster
- Center for Addiction Medicine, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sarah Hickey
- Center for Addiction Medicine, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gladys N Pachas
- Center for Addiction Medicine, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - A Eden Evins
- Center for Addiction Medicine, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jodi M Gilman
- Center for Addiction Medicine, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Abstract
The current review highlights the available research related to cannabis and indicators of physical health in a variety of domains. Various studies have found associations between cannabis use with pulmonary, cardiovascular, gastrointestinal, and endocrine function as well as body mass index and sleep. At this time, more research is needed to understand the influence of cannabis use on physical health, particularly among adolescent samples.
Collapse
Affiliation(s)
- Abigail L Tuvel
- Department of Psychology and Neuroscience, University of Colorado Boulder, 1777 Exposition Drive, Boulder, CO 80301
| | - Evan A Winiger
- Department of Psychiatry, School of Medicine, University of Colorado Anschutz Medical Campus, 1890 N Revere Court, Aurora, CO, 80045
| | - J Megan Ross
- Department of Psychiatry, Division of Addiction Sciences, Treatment and Prevention, University of Colorado Anschutz Medical Campus, 1890 N Revere Court, Aurora, CO, 80045.
| |
Collapse
|
12
|
Thomas PA, Carter GT, Bombardier CH. A scoping review on the effect of cannabis on pain intensity in people with spinal cord injury. J Spinal Cord Med 2022; 45:656-667. [PMID: 33465022 PMCID: PMC9542582 DOI: 10.1080/10790268.2020.1865709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
CONTEXT This scoping review examines the current research on the effect of cannabis upon pain intensity in spinal cord injury (SCI) pain. Chronic pain is a significant secondary condition following SCI, and traditional treatments (e.g. opioids, NSAIDs) are often criticized for providing inadequate relief. As a result, there is increasing interest in and use of cannabis and cannabinoid-based medications as an alternative means of pain control. OBJECTIVE The purpose of this review was to examine the scientific evidence on the effect of cannabis/cannabinoids upon pain intensity in SCI by mapping the current literature. METHODS Two hundred and fifty-two studies were identified by searching electronic databases for articles published through February 2020. In addition, reviewers scanned the reference lists of identified articles and examined clinicaltrials.gov for unpublished data in this area. Title, abstract, and full-text reviews were completed by two independent reviewers. Data extraction was performed by a single reviewer and verified by a second reviewer. RESULTS Six articles covering five treatment studies were included. Studies yielded mixed findings likely due to large variability in methodology, including lack of standardized dosing paradigms, modes of use, and duration of trial. CONCLUSIONS The current quality and level of evidence is insufficient to draw reliable conclusions of the efficacy of cannabis upon SCI-related pain itensity. We identify specific limitations of past studies and present guidelines for future research.Trial registration: ClinicalTrials.gov identifier: Nct01606202..
Collapse
Affiliation(s)
- Pavithra A. Thomas
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA
| | | | - Charles H. Bombardier
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
13
|
Murillo-Rodríguez E, Coronado-Álvarez A, López-Muciño LA, Pastrana-Trejo JC, Viana-Torre G, Barberena JJ, Soriano-Nava DM, García-García F. Neurobiology of dream activity and effects of stimulants on dreams. Curr Top Med Chem 2022; 22:1280-1295. [PMID: 35761491 DOI: 10.2174/1568026622666220627162032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 03/18/2022] [Accepted: 04/11/2022] [Indexed: 11/22/2022]
Abstract
The sleep-wake cycle is the result of the activity of a multiple neurobiological network interaction. Dreaming feature is one interesting sleep phenomena that represents sensorial components, mostly visual perceptions, accompanied with intense emotions. Further complexity has been added to the topic of the neurobiological mechanism of dreams generation by the current data that suggests the influence of drugs on dream generation. Here, we discuss the review on some of the neurobiological mechanism of the regulation of dream activity, with special emphasis on the effects of stimulants on dreaming.
Collapse
Affiliation(s)
- Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group
| | - Astrid Coronado-Álvarez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group
| | - Luis Angel López-Muciño
- Health Sciences Program. Health Sciences Institute. Veracruzana University. Xalapa. Veracruz. Mexico
| | - José Carlos Pastrana-Trejo
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group
| | - Gerardo Viana-Torre
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group
| | - Juan José Barberena
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group.,Escuela de Psicología, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México
| | - Daniela Marcia Soriano-Nava
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group
| | - Fabio García-García
- Intercontinental Neuroscience Research Group.,Health Sciences Program. Health Sciences Institute. Veracruzana University. Xalapa. Veracruz. Mexico
| |
Collapse
|
14
|
Gilman JM, Schuster RM, Potter KW, Schmitt W, Wheeler G, Pachas GN, Hickey S, Cooke ME, Dechert A, Plummer R, Tervo-Clemmens B, Schoenfeld DA, Evins AE. Effect of Medical Marijuana Card Ownership on Pain, Insomnia, and Affective Disorder Symptoms in Adults: A Randomized Clinical Trial. JAMA Netw Open 2022; 5:e222106. [PMID: 35302633 PMCID: PMC8933735 DOI: 10.1001/jamanetworkopen.2022.2106] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/24/2022] [Indexed: 01/30/2023] Open
Abstract
Importance Despite the legalization and widespread use of cannabis products for a variety of medical concerns in the US, there is not yet a strong clinical literature to support such use. The risks and benefits of obtaining a medical marijuana card for common clinical outcomes are largely unknown. Objective To evaluate the effect of obtaining a medical marijuana card on target clinical and cannabis use disorder (CUD) symptoms in adults with a chief concern of chronic pain, insomnia, or anxiety or depressive symptoms. Design, Setting, and Participants This pragmatic, single-site, single-blind randomized clinical trial was conducted in the Greater Boston area from July 1, 2017, to July 31, 2020. Participants were adults aged 18 to 65 years with a chief concern of pain, insomnia, or anxiety or depressive symptoms. Participants were randomized 2:1 to either the immediate card acquisition group (n = 105) or the delayed card acquisition group (n = 81). Randomization was stratified by chief concern, age, and sex. The statistical analysis followed an evaluable population approach. Interventions The immediate card acquisition group was allowed to obtain a medical marijuana card immediately after randomization. The delayed card acquisition group was asked to wait 12 weeks before obtaining a medical marijuana card. All participants could choose cannabis products from a dispensary, the dose, and the frequency of use. Participants could continue their usual medical or psychiatric care. Main Outcomes and Measures Primary outcomes were changes in CUD symptoms, anxiety and depressive symptoms, pain severity, and insomnia symptoms during the trial. A logistic regression model was used to estimate the odds ratio (OR) for CUD diagnosis, and linear models were used for continuous outcomes to estimate the mean difference (MD) in symptom scores. Results A total of 186 participants (mean [SD] age 37.2 [14.4] years; 122 women [65.6%]) were randomized and included in the analyses. Compared with the delayed card acquisition group, the immediate card acquisition group had more CUD symptoms (MD, 0.28; 95% CI, 0.15-0.40; P < .001); fewer self-rated insomnia symptoms (MD, -2.90; 95% CI, -4.31 to -1.51; P < .001); and reported no significant changes in pain severity or anxiety or depressive symptoms. Participants in the immediate card acquisition group also had a higher incidence of CUD during the intervention (17.1% [n = 18] in the immediate card acquisition group vs 8.6% [n = 7] in the delayed card acquisition group; adjusted odds ratio, 2.88; 95% CI, 1.17-7.07; P = .02), particularly those with a chief concern of anxiety or depressive symptoms. Conclusions and Relevance This randomized clinical trial found that immediate acquisition of a medical marijuana card led to a higher incidence and severity of CUD; resulted in no significant improvement in pain, anxiety, or depressive symptoms; and improved self-rating of insomnia symptoms. Further investigation of the benefits of medical marijuana card ownership for insomnia and the risk of CUD are needed, particularly for individuals with anxiety or depressive symptoms. Trial Registration ClinicalTrials.gov Identifier: NCT03224468.
Collapse
Affiliation(s)
- Jodi M. Gilman
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, MGH, Harvard Medical School, Charlestown
| | - Randi M. Schuster
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Kevin W. Potter
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - William Schmitt
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, MGH, Harvard Medical School, Charlestown
| | - Grace Wheeler
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, MGH, Harvard Medical School, Charlestown
| | - Gladys N. Pachas
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Sarah Hickey
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
| | - Megan E. Cooke
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Alyson Dechert
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, MGH, Harvard Medical School, Charlestown
| | - Rachel Plummer
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, MGH, Harvard Medical School, Charlestown
| | - Brenden Tervo-Clemmens
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - David A. Schoenfeld
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
- Department of Biostatistics, MGH, Boston
| | - A. Eden Evins
- Department of Psychiatry, Massachusetts General Hospital (MGH), Boston
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Maddison KJ, Kosky C, Walsh JH. Is There a Place for Medicinal Cannabis in Treating Patients with Sleep Disorders? What We Know so Far. Nat Sci Sleep 2022; 14:957-968. [PMID: 35611178 PMCID: PMC9124464 DOI: 10.2147/nss.s340949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/06/2022] [Indexed: 02/02/2023] Open
Abstract
The legalization of cannabis for medicinal, and in some countries, recreational, purposes in addition to growth in the cannabis industry has meant that cannabis use and interest in the area has increased rapidly over the past 20 years. Treatment of poor sleep and sleep disorders are two of the most common reasons for the current use of medicinal cannabis. However, evidence for the role of medical cannabis in the treatment of sleep disorders has not been clearly established, thus making it challenging for clinicians to make evidence-based decisions regarding efficacy and safety. This narrative review summarizes the highest quality clinical evidence currently available in relation to the use of medicinal cannabis for the treatment of sleep disorders including insomnia, obstructive sleep apnea, restless legs syndrome, rapid eye movement sleep behavior disorder, nightmare disorder and narcolepsy. A summary of the effect of cannabis on sleep quality and architecture is also presented. Currently, there is insufficient evidence to support the routine use of medicinal cannabis as an effective and safe treatment option for any sleep disorder. Nevertheless, emerging evidence is promising and warrants further investigation using standardized cannabinoid products and validated quantitative measurement techniques.
Collapse
Affiliation(s)
- Kathleen J Maddison
- West Australian Sleep Disorders Research Institute, Department of Pulmonary Physiology & Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,Centre for Sleep Science, University of Western Australia, Perth, Western Australia, Australia
| | - Christopher Kosky
- West Australian Sleep Disorders Research Institute, Department of Pulmonary Physiology & Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Jennifer H Walsh
- West Australian Sleep Disorders Research Institute, Department of Pulmonary Physiology & Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,Centre for Sleep Science, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
16
|
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
|
17
|
Rao A, Ebelt P, Mallard A, Briskey D. Palmitoylethanolamide for sleep disturbance. A double-blind, randomised, placebo-controlled interventional study. SLEEP SCIENCE AND PRACTICE 2021; 5:12. [PMID: 34522787 PMCID: PMC8428962 DOI: 10.1186/s41606-021-00065-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/12/2021] [Indexed: 11/12/2022] Open
Abstract
Background Sleep is essential for wellbeing, yet sleep disturbance is a common problem linked to a wide range of health conditions. Palmitoylethanolamide (PEA) is an endogenous fatty acid amide proposed to promote better sleep via potential interaction with the endocannabinoid system. Methods This double-blind, randomised study on 103 adults compared the efficacy and tolerability of 8 weeks of daily supplemented PEA formulation (350 mg Levagen + ®) to a placebo. Sleep quality and quantity were measured using wrist actigraphy, a sleep diary and questionnaires. Results At week 8, PEA supplementation reduced sleep onset latency, time to feel completely awake and improved cognition on waking. After 8 weeks, both groups improved their sleep quality and quantity scores similarly. There was no difference between groups at baseline or week 8 for sleep quantity or quality as measured from actigraphy or sleep diaries. Conclusion These findings support PEA as a potential sleeping aid capable of reducing sleep onset time and improving cognition on waking. Trial registration Australian
New Zealand Clinical Trials Registry ACTRN12618001339246. Registered 9th
August 2018.
Collapse
Affiliation(s)
- Amanda Rao
- RDC Clinical, Brisbane, 4006 Australia.,University of Sydney, School of Medicine, Sydney, Australia
| | | | - Alistair Mallard
- Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - David Briskey
- RDC Clinical, Brisbane, 4006 Australia.,School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, Australia
| |
Collapse
|
18
|
Murillo-Rodríguez E, Arankowsky-Sandoval G, Budde H, Imperatori C, Machado S, Yamamoto T, Yadollahpour A, Torterolo P. In vivo brain levels of acetylcholine and 5-hydroxytryptamine after oleoylethanolamide or palmitoylethanolamide administrations are mediated by PPARα engagement. Eur J Neurosci 2021; 54:5932-5950. [PMID: 34396611 DOI: 10.1111/ejn.15409] [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: 05/18/2021] [Revised: 07/06/2021] [Accepted: 07/25/2021] [Indexed: 11/29/2022]
Abstract
The peroxisome proliferator-activated receptor alpha (PPARα) is a nuclear receptor that has been linked to the modulation of several physiological functions, including the sleep-wake cycle. The PPARα recognizes as endogenous ligands the lipids oleoylethanolamide (OEA) and palmitoylethanolamide (PEA), which in turn, if systemically injected, they exert wake-promoting effects. Moreover, the activation of PPARα by the administration of OEA or PEA increases the extracellular contents of neurotransmitters linked to the control of wakefulness; however, the role of PPARα activated by OEA or PEA on additional biochemicals related to waking regulation, such as acetylcholine (ACh) and 5-hydroxytryptamine (5-HT), has not been fully studied. Here, we have investigated the effects of treatments of OEA or PEA on the contents of ACh and 5-HT by using in vivo microdialysis techniques coupled to HPLC means. For this purpose, OEA or PEA were systemically injected (5, 10 or 30 mg/kg; i.p.), and the levels of ACh and 5-HT were collected from the basal forebrain, a wake-related brain area. These pharmacological treatments significantly increased the contents of ACh and 5-HT as determined by HPLC procedures. Interestingly, PPARα antagonist MK-886 (30 mg/kg; i.p.) injected before the treatments of OEA or PEA blocked these outcomes. Our data suggest that the activation of PPARα by OEA or PEA produces significant changes on ACh and 5-HT levels measured from the basal forebrain and support the conclusion that PPARα is a suitable molecular element involved in the regulation of wake-related neurotransmitters.
Collapse
Affiliation(s)
- Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mérida, Yucatán, Mexico.,Intercontinental Neuroscience Research Group
| | - Gloria Arankowsky-Sandoval
- Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Henning Budde
- Intercontinental Neuroscience Research Group.,Institute for Systems Medicine, Faculty of Human Sciences, MSH Medical School Hamburg, Hamburg, Germany
| | - Claudio Imperatori
- Intercontinental Neuroscience Research Group.,Cognitive and Clinical Psychology Laboratory, Department of Human Science, European University of Rome, Rome, Italy
| | - Sérgio Machado
- Intercontinental Neuroscience Research Group.,Department of Sports Methods and Techniques, Federal University of Santa Maria, Santa Maria, Brazil.,Laboratory of Physical Activity Neuroscience, Neurodiversity Institute, Queimados, Brazil
| | - Tetsuya Yamamoto
- Intercontinental Neuroscience Research Group.,Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Ali Yadollahpour
- Intercontinental Neuroscience Research Group.,Department of Psychology, University of Sheffield, Sheffield, UK
| | - Pablo Torterolo
- Intercontinental Neuroscience Research Group.,Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
19
|
Edwards D, Filbey FM. Are Sweet Dreams Made of These? Understanding the Relationship Between Sleep and Cannabis Use. Cannabis Cannabinoid Res 2021; 6:462-473. [PMID: 34143657 DOI: 10.1089/can.2020.0174] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Introduction: There is widespread literature on the interaction between cannabis use and sleep, yet the mechanisms that underlie this relationship are not well understood. Several factors lead to inconsistencies in this relationship suggesting a nuanced interaction between cannabis and sleep. An important question that remains to be addressed is the temporal relationship between disrupted sleep and cannabis use. This literature review summarizes the existing literature on the association between disrupted sleep and cannabis toward the goal of addressing the question of the chronology of these reported effects. Materials and Methods: We conducted a review of the literature using PubMed to summarize current knowledge on the association between cannabis use and sleep in humans. Results: We identified 31 studies on the association of cannabis use and sleep. The findings from these studies were mixed. Cannabis was associated with a variety of impacts on sleep ranging from beneficial effects, such as reduced sleep-onset latency, to negative outcomes, such as reduced sleep duration and suppressed rapid eye movement oscillations. The chronology of the interaction of cannabis and sleep was unclear, although much of the current literature focus on factors that modulate how cannabis impairs sleep after initial use. Conclusion: There was sufficient evidence to suggest that cannabis use alters circadian rhythms, and hence, negatively impacts sleep. The current literature is largely from studies utilizing self-report measures of sleep; thus, objective measures of sleep are needed. In addition, although there were no empirical studies on the temporal relationship between cannabis use and sleep, the majority of the literature focused on characterizing sleep impairment after cannabis use.
Collapse
Affiliation(s)
- Dalton Edwards
- Center for BrainHealth, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas, USA
| | - Francesca M Filbey
- Center for BrainHealth, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas, USA
| |
Collapse
|
20
|
Ni R, Bhandari S, Mitchell PR, Suarez G, Patel NB, Lamb K, Bisht KS, Merkler DJ. Synthesis, Quantification, and Characterization of Fatty Acid Amides from In Vitro and In Vivo Sources. Molecules 2021; 26:molecules26092543. [PMID: 33925418 PMCID: PMC8123904 DOI: 10.3390/molecules26092543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 01/01/2023] Open
Abstract
Fatty acid amides are a diverse family of underappreciated, biologically occurring lipids. Herein, the methods for the chemical synthesis and subsequent characterization of specific members of the fatty acid amide family are described. The synthetically prepared fatty acid amides and those obtained commercially are used as standards for the characterization and quantification of the fatty acid amides produced by biological systems, a fatty acid amidome. The fatty acid amidomes from mouse N18TG2 cells, sheep choroid plexus cells, Drosophila melanogaster, Bombyx mori, Apis mellifera, and Tribolium castaneum are presented.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kirpal S. Bisht
- Correspondence: (K.S.B.); (D.J.M.); Tel.: +1-813-974-0350 (K.S.B.); +1-813-974-3579 (D.J.M.)
| | - David J. Merkler
- Correspondence: (K.S.B.); (D.J.M.); Tel.: +1-813-974-0350 (K.S.B.); +1-813-974-3579 (D.J.M.)
| |
Collapse
|
21
|
Wilson NL, Peterson SN, Ellis RJ. Cannabis and the Gut-Brain Axis Communication in HIV Infection. Cannabis Cannabinoid Res 2021; 6:92-104. [PMID: 33912676 PMCID: PMC8064951 DOI: 10.1089/can.2020.0037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
People living with HIV infection (PWH) disclose that cannabis is an effective strategy for alleviating symptoms associated with HIV disease. However, some medical providers feel ill-informed to engage in evidence-based conversations. HIV leads to alterations in the gut microbiome, gut-brain axis signaling, and chronic inflammation. The endocannabinoid system regulates homeostasis of multiple organ systems. When deficient, dysregulation of the gut-brain axis can result in chronic inflammation and neuroinflammation. Cannabis along with the naturally occurring endocannabinoids has antioxidant and anti-inflammatory properties that can support healing and restoration as an adjunctive therapy. The purpose of this literature review is to report the physiologic mechanisms that occur in the pathology of HIV and discuss potential benefits of cannabinoids in supporting health and reducing the negative effects of comorbidities in PWH.
Collapse
Affiliation(s)
- Natalie L. Wilson
- Department of Community Health Systems, School of Nursing, University of California, San Francisco, San Francisco, California, USA
| | - Scott N. Peterson
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Ronald J. Ellis
- Departments of Neurosciences and Psychiatry, University of California, San Diego, San Diego, California, USA
| |
Collapse
|
22
|
Winiger EA, Ellingson JM, Morrison CL, Corley RP, Pasman JA, Wall TL, Hopfer CJ, Hewitt JK. Sleep deficits and cannabis use behaviors: an analysis of shared genetics using linkage disequilibrium score regression and polygenic risk prediction. Sleep 2021; 44:zsaa188. [PMID: 32935850 PMCID: PMC7953210 DOI: 10.1093/sleep/zsaa188] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
STUDY OBJECTIVES Estimate the genetic relationship of cannabis use with sleep deficits and an eveningness chronotype. METHODS We used linkage disequilibrium score regression (LDSC) to analyze genetic correlations between sleep deficits and cannabis use behaviors. Secondly, we generated sleep deficit polygenic risk score (PRS) and estimated their ability to predict cannabis use behaviors using linear and logistic regression. Summary statistics came from existing genome-wide association studies of European ancestry that were focused on sleep duration, insomnia, chronotype, lifetime cannabis use, and cannabis use disorder (CUD). A target sample for PRS prediction consisted of high-risk participants and participants from twin/family community-based studies (European ancestry; n = 760, male = 64%; mean age = 26.78 years). Target data consisted of self-reported sleep (sleep duration, feeling tired, and taking naps) and cannabis use behaviors (lifetime ever use, number of lifetime uses, past 180-day use, age of first use, and lifetime CUD symptoms). RESULTS Significant genetic correlation between lifetime cannabis use and an eveningness chronotype (rG = 0.24, p < 0.001), as well as between CUD and both short sleep duration (<7 h; rG = 0.23, p = 0.017) and insomnia (rG = 0.20, p = 0.020). Insomnia PRS predicted earlier age of first cannabis use (OR = 0.92, p = 0.036) and increased lifetime CUD symptom count (OR = 1.09, p = 0.012). CONCLUSION Cannabis use is genetically associated with both sleep deficits and an eveningness chronotype, suggesting that there are genes that predispose individuals to both cannabis use and sleep deficits.
Collapse
Affiliation(s)
- Evan A Winiger
- Institute for Behavioral Genetics, University of Colorado Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado Boulder, CO
| | - Jarrod M Ellingson
- Institute for Behavioral Genetics, University of Colorado Boulder, CO
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO
| | - Claire L Morrison
- Institute for Behavioral Genetics, University of Colorado Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado Boulder, CO
| | - Robin P Corley
- Institute for Behavioral Genetics, University of Colorado Boulder, CO
| | - Joëlle A Pasman
- Behavioural Science Institute, Radboud University Nijmegen, Amsterdam, The Netherlands
| | - Tamara L Wall
- Department of Psychiatry, University of California, San Diego, CA
| | - Christian J Hopfer
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO
| | - John K Hewitt
- Institute for Behavioral Genetics, University of Colorado Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado Boulder, CO
| |
Collapse
|
23
|
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]
|
24
|
Mediating Effect of Perceived Stress on the Association between Physical Activity and Sleep Quality among Chinese College Students. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18010289. [PMID: 33401720 PMCID: PMC7795525 DOI: 10.3390/ijerph18010289] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND While physical activity has been reported to positively affect stress and sleep quality, less is known about the potential relationships among them. The present study aimed to investigate the mediating effect of stress on the association between physical activity and sleep quality in Chinese college students, after controlling for age, nationality, and tobacco and alcohol use. PARTICIPANTS The sample comprised 6973 college students representing three Chinese universities. METHODS Physical activity, perceived stress, and sleep quality were respectively measured using the International Physical Activity Questionnaire-Short Form (IPAQ-SF), Perceived Stress Scale-10 Items (PSS-10), and Pittsburgh Sleep Quality Index (PSQI). RESULTS Mediating effects of perceived stress on the association between physical activity and sleep quality were observed in males and females, with 42.4% (partial mediating effect) and 306.3% (complete mediating effect) as percentages of mediation, respectively. CONCLUSION The results of this study may provide some suggestions that physical activity could improve sleep by aiding individuals in coping with stress and indicate that stress management might be an effective non-pharmaceutical therapy for sleep improvement.
Collapse
|
25
|
Ek J, Jacobs W, Kaylor B, McCall WV. Addiction and Sleep Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1297:163-171. [PMID: 33537944 DOI: 10.1007/978-3-030-61663-2_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Shared neurophysiology of addiction and sleep disorders results in a bidirectional interplay. Diagnosing and treating primary sleep disorders, particularly in adolescents, can prevent the development of addiction in susceptible individuals. Addressing sleep issues in early recovery, and throughout maintenance, can prevent relapse. Cannabis use for insomnia shows mixed results; assisting with onset sleep latency in early use, this subsides with chronic use and holds addiction risk. Insomnia is a primary complaint of cannabis withdrawal syndrome and a primary cause of relapse in cannabis use disorder. An ideal sleep aid would prevent relapse and have low abuse potential. Pharmaceutical and behavioral options include suvorexant, mirtazapine, trazodone, and aerobic exercise, but clinical trials are lacking to demonstrate efficacy.
Collapse
Affiliation(s)
- Jonathan Ek
- Division of Addiction Medicine, Augusta University-Medical College of Georgia, Augusta, GA, USA.
| | - William Jacobs
- Division of Addiction Medicine, Augusta University-Medical College of Georgia, Augusta, GA, USA
| | - Brett Kaylor
- Division of Addiction Medicine, Augusta University-Medical College of Georgia, Augusta, GA, USA
| | - W Vaughn McCall
- Division of Addiction Medicine, Augusta University-Medical College of Georgia, Augusta, GA, USA
| |
Collapse
|
26
|
Abstract
The sleep-wake cycle is a complex process that includes wake (W), non-rapid-eye-movement (NREM) and rapid-eye-movement (REM) sleep. Each phase is regulated by specialized brain structures that, by means of different neurotransmitters, maintain the constant expression of the sleep-wake cycle. Molecules like orexin, serotonin, noradrenaline, histamine, for waking; GABA, adenosine, prostaglandins, for NREM sleep and acetylcholine and glutamate for REM sleep, among other molecules are responsible for the expression and maintenance of each phase. When the endocannabinoid system was being described for the first time, almost three decades ago, oleamide's sleep promoting properties were highlighted. Nowadays, enough evidence has been cumulated to support the endocannabinoid system role in the sleep-wake cycle regulation. The endocannabinoids oleamide anandamide, and 2-arachidonylglycerol promote NREM and/or REM sleep via the CB1R, thereby making this system a target to treat sleep disorders, such as insomnia.
Collapse
|
27
|
Abstract
The American Thoracic Society Core Curriculum updates clinicians annually in adult and pediatric pulmonary disease, medical critical care, and sleep medicine, in a 3–4-year recurring cycle of topics. These topics will be presented at the 2020 Virtual Conference. Below is the adult sleep medicine core that includes topics pertinent to sleep-disordered breathing and insomnia.
Collapse
|
28
|
Cannabinoids in the Treatment of Insomnia Disorder: A Systematic Review and Meta-Analysis. CNS Drugs 2020; 34:1217-1228. [PMID: 33244728 DOI: 10.1007/s40263-020-00773-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/01/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Insomnia is associated with significant comorbidity, disability and impact on quality of life and, despite advances in pharmacotherapy and psychotherapy, remains a significant burden to society. Cannabinoids are gaining acceptance for use as medicines in the treatment of insomnia disorder. OBJECTIVE We conducted a systematic review and meta-analysis to evaluate the efficacy of cannabinoids in the treatment of insomnia disorder. METHODS We performed a systematic review of the PubMed, Cochrane Library, MEDLINE, and Cumulative Index to Nursing and Allied Health Literature Complete databases from inception to 5 December 2019, and again prior to data abstraction, for studies of cannabis-based products for the treatment of insomnia disorder in adults. Inclusion criteria were (1) clinical studies, (2) participants aged ≥ 18 years, (3) insomnia disorder either formally diagnosed against contemporaneous diagnostic criteria or quantified with validated instruments and (4) compared cannabis-based products with the standard of care, placebo or a sedative. No language restrictions were imposed. Non-primary research, animal studies and studies of cannabis-induced insomnia were excluded. Risk of bias was assessed using the RoB 2 tool for randomised controlled trials (RCTs) and Risk of Bias in Non-randomized Studies-of Interventions (ROBINS-I) tool for non-randomized trials. Heterogeneity was assessed with the I2 statistic. RESULTS A total of five studies (two RCTs and three non-randomised studies) with 219 study participants were included, of which three could be combined. The three non-randomised studies contributed data on the Pittsburgh Sleep Quality Index Questionnaire score, showing a favourable effect of cannabinoids at ≤ 4 weeks of follow-up (mean difference - 1.89 [95% confidence interval {CI} - 2.68 to - 1.10]; n = 176) and at 8 weeks of follow-up (mean difference - 2.41 [95% CI - 3.36 to - 1.46]; n = 166). One double-blind crossover RCT (n = 32) reported that, compared with amitriptyline, nabilone-a synthetic analogue to tetrahydrocannabinol (THC)-improved Insomnia Severity Index scores after 2 weeks of treatment (adjusted difference - 3.25 [95% CI - 5.26 to - 1.24]) and resulted in a more restful sleep as a sub-measure of the Leeds Sleep Evaluation Questionnaire (LSEQ) (difference 0.48 [95% CI 0.01-0.95]) but with no effect on overall sleep quality as measured by the LSEQ. In a single ascending-dose RCT (n = 9), THC reduced sleep-onset latency compared with placebo at 10 mg, 20 mg and 30 mg doses (mean difference - 43.00 min [95% CI - 82.76 to - 3.24], - 62.00 [95% CI - 103.60 to - 20.40] and - 54.00 [95% CI - 103.93 to - 4.07], respectively). All the included studies were assessed as poor quality, mainly due to small sample sizes, short treatment periods, uncertain clinical significance and high risk of bias. CONCLUSIONS Few studies have examined the efficacy of cannabinoids in the treatment of insomnia disorder. Despite some possible signals for efficacy, the heterogeneity of participants, interventions, efficacy outcomes and results, and the high risk of bias across included trials, do not reliably inform evidence-based practice. This review highlights shortcomings in the existing literature, including lack of diagnostic clarity, poorly defined participant groups, non-standardised interventions and studies of inappropriate design, duration and power to detect clinically meaningful outcomes. Further research in the form of high-quality RCTs are required before drawing any conclusions about the efficacy of cannabinoids in the treatment of insomnia disorder. TRIAL REGISTRATION PROSPERO registration number, CRD42020161043.
Collapse
|
29
|
Sznitman SR, Vulfsons S, Meiri D, Weinstein G. Medical cannabis and insomnia in older adults with chronic pain: a cross-sectional study. BMJ Support Palliat Care 2020; 10:415-420. [PMID: 31959585 DOI: 10.1136/bmjspcare-2019-001938] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Medical cannabis (MC) is increasingly being used for treatment of chronic pain symptoms. Among patients there is also a growing preference for the use of MC to manage sleep problems. The aim of the current study was to examine the associations between use of whole plant cannabis and sleep problems among chronic pain patients. METHODS A total of 128 individuals with chronic pain over the age of 50 years were recruited from the Rambam Institute for Pain Medicine in Haifa, Israel. Of them, 66 were MC users and 62 were non-users. Regression models tested the differences in sleep problems between the two groups. Furthermore, Pearson correlations between MC use measures (dose, length and frequency of use, number of strains used, tetrahydrocannabinol/cannabidiol levels) and sleep problems were assessed among MC users. RESULTS After adjustment for age, sex, pain level and use of sleep and anti-depressant medications, MC use was associated with less problems with waking up at night compared with non-MC use. No group differences were found for problems with falling asleep or waking up early without managing to fall back asleep. Frequent MC use was associated with more problems waking up at night and falling asleep. CONCLUSIONS MC use may have an overall positive effect on maintaining sleep throughout the night in chronic pain patients. At the same time, tolerance towards potential sleep-inducing properties of MC may occur with frequent use. More research based on randomised control trials and other longitudinal designs is warranted.
Collapse
Affiliation(s)
- Sharon R Sznitman
- School of Public Health, University of Haifa Faculty of Social Welfare and Health Sciences, Haifa, Israel
| | - Simon Vulfsons
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
- Institute of Pain Medicine, Rambam Health Care Campus, Haifa, Israel
| | - David Meiri
- Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
| | - Galit Weinstein
- School of Public Health, University of Haifa Faculty of Social Welfare and Health Sciences, Haifa, Israel
| |
Collapse
|
30
|
Boyer C, Cussonneau L, Brun C, Deval C, Pais de Barros JP, Chanon S, Bernoud-Hubac N, Daira P, Evans AL, Arnemo JM, Swenson JE, Gauquelin-Koch G, Simon C, Blanc S, Combaret L, Bertile F, Lefai E. Specific shifts in the endocannabinoid system in hibernating brown bears. Front Zool 2020; 17:35. [PMID: 33292302 PMCID: PMC7681968 DOI: 10.1186/s12983-020-00380-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/20/2020] [Indexed: 01/30/2023] Open
Abstract
In small hibernators, global downregulation of the endocannabinoid system (ECS), which is involved in modulating neuronal signaling, feeding behavior, energy metabolism, and circannual rhythms, has been reported to possibly drive physiological adaptation to the hibernating state. In hibernating brown bears (Ursus arctos), we hypothesized that beyond an overall suppression of the ECS, seasonal shift in endocannabinoids compounds could be linked to bear’s peculiar features that include hibernation without arousal episodes and capacity to react to external disturbance. We explored circulating lipids in serum and the ECS in plasma and metabolically active tissues in free-ranging subadult Scandinavian brown bears when both active and hibernating. In winter bear serum, in addition to a 2-fold increase in total fatty acid concentration, we found significant changes in relative proportions of circulating fatty acids, such as a 2-fold increase in docosahexaenoic acid C22:6 n-3 and a decrease in arachidonic acid C20:4 n-6. In adipose and muscle tissues of hibernating bears, we found significant lower concentrations of 2-arachidonoylglycerol (2-AG), a major ligand of cannabinoid receptors 1 (CB1) and 2 (CB2). Lower mRNA level for genes encoding CB1 and CB2 were also found in winter muscle and adipose tissue, respectively. The observed reduction in ECS tone may promote fatty acid mobilization from body fat stores, and favor carbohydrate metabolism in skeletal muscle of hibernating bears. Additionally, high circulating level of the endocannabinoid-like compound N-oleoylethanolamide (OEA) in winter could favor lipolysis and fatty acid oxidation in peripheral tissues. We also speculated on a role of OEA in the conservation of an anorexigenic signal and in the maintenance of torpor during hibernation, while sustaining the capacity of bears to sense stimuli from the environment.
Collapse
Affiliation(s)
- Christian Boyer
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Laura Cussonneau
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Charlotte Brun
- Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
| | - Christiane Deval
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | | | - Stéphanie Chanon
- Université de Lyon, INSERM, INRAE, INSA, Functional Lipidomic Plateform, Lyon, France
| | | | - Patricia Daira
- Université de Lyon, INSERM, INRAE, INSA, Functional Lipidomic Plateform, Lyon, France
| | - Alina L Evans
- Department of Forestry and Wildlife Management, Inland Norway University of Applied Sciences, Campus Evenstad, NO-2480, Koppang, Norway
| | - Jon M Arnemo
- Department of Forestry and Wildlife Management, Inland Norway University of Applied Sciences, Campus Evenstad, NO-2480, Koppang, Norway.,Department of Wildlife, Fish, and Environmental Studies, Swedish University of Agricultural Sciences, SE-901 83, Umeå, Sweden
| | - Jon E Swenson
- Faculty of Environmental Sciences and Natural Resource Management, Norwegian University of Life Sciences, NO-1432, Ås, Norway
| | | | - Chantal Simon
- Université de Lyon, INSERM, INRAE, INSA, Functional Lipidomic Plateform, Lyon, France
| | - Stéphane Blanc
- Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
| | - Lydie Combaret
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Fabrice Bertile
- Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
| | - Etienne Lefai
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France.
| |
Collapse
|
31
|
Fasciani I, Petragnano F, Aloisi G, Marampon F, Carli M, Scarselli M, Maggio R, Rossi M. Allosteric Modulators of G Protein-Coupled Dopamine and Serotonin Receptors: A New Class of Atypical Antipsychotics. Pharmaceuticals (Basel) 2020; 13:ph13110388. [PMID: 33202534 PMCID: PMC7696972 DOI: 10.3390/ph13110388] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/23/2022] Open
Abstract
Schizophrenia was first described by Emil Krapelin in the 19th century as one of the major mental illnesses causing disability worldwide. Since the introduction of chlorpromazine in 1952, strategies aimed at modifying the activity of dopamine receptors have played a major role for the treatment of schizophrenia. The introduction of atypical antipsychotics with clozapine broadened the range of potential targets for the treatment of this psychiatric disease, as they also modify the activity of the serotoninergic receptors. Interestingly, all marketed drugs for schizophrenia bind to the orthosteric binding pocket of the receptor as competitive antagonists or partial agonists. In recent years, a strong effort to develop allosteric modulators as potential therapeutic agents for schizophrenia was made, mainly for the several advantages in their use. In particular, the allosteric binding sites are topographically distinct from the orthosteric pockets, and thus drugs targeting these sites have a higher degree of receptor subunit specificity. Moreover, “pure” allosteric modulators maintain the temporal and spatial fidelity of native orthosteric ligand. Furthermore, allosteric modulators have a “ceiling effect”, and their modulatory effect is saturated above certain concentrations. In this review, we summarize the progresses made in the identification of allosteric drugs for dopamine and serotonin receptors, which could lead to a new generation of atypical antipsychotics with a better profile, especially in terms of reduced side effects.
Collapse
Affiliation(s)
- Irene Fasciani
- Department of Biotechnological and Applied Clinical Sciences, University of l’Aquila, 67100 L’Aquila, Italy; (I.F.); (F.P.); (G.A.)
| | - Francesco Petragnano
- Department of Biotechnological and Applied Clinical Sciences, University of l’Aquila, 67100 L’Aquila, Italy; (I.F.); (F.P.); (G.A.)
| | - Gabriella Aloisi
- Department of Biotechnological and Applied Clinical Sciences, University of l’Aquila, 67100 L’Aquila, Italy; (I.F.); (F.P.); (G.A.)
| | - Francesco Marampon
- Department of Radiotherapy, “Sapienza” University of Rome, Policlinico Umberto I, 00161 Rome, Italy;
| | - Marco Carli
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (M.C.); (M.S.)
| | - Marco Scarselli
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (M.C.); (M.S.)
| | - Roberto Maggio
- Department of Biotechnological and Applied Clinical Sciences, University of l’Aquila, 67100 L’Aquila, Italy; (I.F.); (F.P.); (G.A.)
- Correspondence:
| | - Mario Rossi
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, UK;
| |
Collapse
|
32
|
|
33
|
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
|
34
|
Potvin S, Mahrouche L, Assaf R, Chicoine M, Giguère CÉ, Furtos A, Godbout R. Peripheral Endogenous Cannabinoid Levels Are Increased in Schizophrenia Patients Evaluated in a Psychiatric Emergency Setting. Front Psychiatry 2020; 11:628. [PMID: 32695035 PMCID: PMC7338686 DOI: 10.3389/fpsyt.2020.00628] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/16/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The endogenous cannabinoid system mediates the psychoactive effects of cannabis in the brain. It has been argued that this system may play a key role in the pathophysiology of schizophrenia. While some studies have consistently shown that the levels of anandamide, an endogenous cannabinoid ligand, are increased in the cerebrospinal fluid of schizophrenia patients, inconsistent results have been observed in studies measuring anandamide levels in the periphery. Here, we sought to determine if the assessment of peripheral anandamide levels in patients evaluated in a psychiatric emergency setting would show robust increases. METHODS One hundred seven patients with a schizophrenia-spectrum disorder from the psychiatric emergency settings of the Institut Universitaire en Santé Mentale de Montréal and 36 healthy volunteers were included in the study. A subsample of thirty patients were assessed at two time points: at the emergency and at their discharge from the hospital. Anxious and depressive symptoms, sleep and substance use were assessed using self-report questionnaires. In addition to anandamide, the levels of oleoylethanolamide (OEA), an anorexigenic fatty-acid ethanolamide, were also measured, since the prevalence of the metabolic syndrome is increased in schizophrenia. Plasma levels of anandamide and OEA were measured using liquid chromatography and mass spectrometry. RESULTS Plasma anandamide and OEA levels were significantly increased in schizophrenia patients, relative to controls (Cohen's d=1.0 and 0.5, respectively). Between-group differences remained significant after controlling for metabolic measures. No differences were observed between schizophrenia patients with and without a comorbid substance use disorder at baseline. Importantly, the levels of both endocannabinoids significantly decreased after discharge from the emergency setting. CONCLUSION The current results add to the growing body of evidence of endocannabinoid alterations in schizophrenia. The strong elevation of plasma anandamide levels in schizophrenia patients assessed in the psychiatric emergency setting suggests that anandamide and OEA area potential biomarkers of the psychological turmoil associated with this context.
Collapse
Affiliation(s)
- Stéphane Potvin
- Department of Psychiatry, Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montreal, QC, Canada
- Department of Psychiatry, University of Montreal, Montreal, QC, Canada
| | - Louiza Mahrouche
- Department of Chemistry, University of Montreal, Montreal, QC, Canada
| | - Roxane Assaf
- Department of Psychiatry, Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montreal, QC, Canada
- Department of Psychiatry, University of Montreal, Montreal, QC, Canada
| | - Marjolaine Chicoine
- Sleep Laboratory and Clinic, CIUSSS du Nord-de-l'Île-de-Montréal, Hôpital en santé mentale Rivière-des-Prairies, Montréal, QC, Canada
| | - Charles-Édouard Giguère
- Department of Psychiatry, Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montreal, QC, Canada
| | - Alexandra Furtos
- Department of Chemistry, University of Montreal, Montreal, QC, Canada
| | - Roger Godbout
- Department of Psychiatry, University of Montreal, Montreal, QC, Canada
- Sleep Laboratory and Clinic, CIUSSS du Nord-de-l'Île-de-Montréal, Hôpital en santé mentale Rivière-des-Prairies, Montréal, QC, Canada
| |
Collapse
|
35
|
Winiger EA, Huggett SB, Hatoum AS, Friedman NP, Drake CL, Wright KP, Hewitt JK. Onset of regular cannabis use and young adult insomnia: an analysis of shared genetic liability. Sleep 2020; 43:zsz293. [PMID: 31855253 PMCID: PMC7368342 DOI: 10.1093/sleep/zsz293] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/22/2019] [Indexed: 01/13/2023] Open
Abstract
STUDY OBJECTIVES Estimate the genetic and environmental influences on the relationship between onset of regular cannabis use and young adult insomnia. METHODS In a population-based twin cohort of 1882 twins (56% female, mean age = 22.99, SD = 2.97) we explored the genetic/environmental etiology of the relationship between onset of regular cannabis use and insomnia-related outcomes via multivariate twin models. RESULTS Controlling for sex, current depression symptoms, and prior diagnosis of an anxiety or depression disorder, adult twins who reported early onset for regular cannabis use (age 17 or younger) were more likely to have insomnia (β = 0.07, p = 0.024) and insomnia with short sleep on weekdays (β = 0.08, p = 0.003) as young adults. We found significant genetic contributions for the onset of regular cannabis use (a2 = 76%, p < 0.001), insomnia (a2 = 44%, p < 0.001), and insomnia with short sleep on weekdays (a2 = 37%, p < 0.001). We found significant genetic correlations between onset of regular use and both insomnia (rA = 0.20, p = 0.047) and insomnia with short sleep on weekdays (rA = 0.25, p = 0.008) but no significant environmental associations between these traits. CONCLUSIONS We found common genetic liabilities for early onset of regular cannabis use and insomnia, implying pleiotropic influences of genes on both traits.
Collapse
Affiliation(s)
- Evan A Winiger
- Institute for Behavioral Genetics, University of Colorado - Boulder, Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado - Boulder, Boulder, CO
| | - Spencer B Huggett
- Institute for Behavioral Genetics, University of Colorado - Boulder, Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado - Boulder, Boulder, CO
| | - Alexander S Hatoum
- Institute for Behavioral Genetics, University of Colorado - Boulder, Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado - Boulder, Boulder, CO
| | - Naomi P Friedman
- Institute for Behavioral Genetics, University of Colorado - Boulder, Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado - Boulder, Boulder, CO
| | | | - Kenneth P Wright
- Department of Integrative Physiology, University of Colorado - Boulder, Boulder, CO
| | - John K Hewitt
- Institute for Behavioral Genetics, University of Colorado - Boulder, Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado - Boulder, Boulder, CO
| |
Collapse
|
36
|
Macías-Triana L, Romero-Cordero K, Tatum-Kuri A, Vera-Barrón A, Millán-Aldaco D, Arankowsky-Sandoval G, Piomelli D, Murillo-Rodríguez E. Exposure to the cannabinoid agonist WIN 55, 212–2 in adolescent rats causes sleep alterations that persist until adulthood. Eur J Pharmacol 2020; 874:172911. [DOI: 10.1016/j.ejphar.2020.172911] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/10/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022]
|
37
|
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
|
38
|
Romigi A, Bari M, Liguori C, Izzi F, Rapino C, Nuccetelli M, Battista N, Bernardini S, Centonze D, Mercuri NB, Placidi F, Maccarrone M. CSF Levels of the Endocannabinoid Anandamide are Reduced in Patients with Untreated Narcolepsy Type 1: A Pilot Study. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:142-147. [PMID: 32148204 DOI: 10.2174/1871527319666200309115602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/13/2020] [Accepted: 01/21/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Endocannabinoids (ECs) modulate both excitatory and inhibitory components in the CNS. There is a growing body of evidence that shows ECs influence both hypothalamic orexinergic and histaminergic neurons involved in narcolepsy physiopathology. Therefore, ECs may influence sleep and sleep-wake cycle. OBJECTIVE To evaluate EC levels in the CSF of untreated narcoleptic patients to test whether ECs are dysregulated in Narcolepsy Type 1 (NT1) and Type 2 (NT2). METHODS We compared CSF Anandamide (AEA), 2-Arachidonoylglycerol (2-AG) and orexin in narcoleptic drug-naïve patients and in a sample of healthy subjects. RESULTS We compared NT1 (n=6), NT2 (n=6), and healthy controls (n=6). We found significantly reduced AEA levels in NT1 patients compared to both NT2 and controls. No differences were found between AEA levels in NT2 versus controls and between 2-AG levels in all groups, although a trend toward a decrease in NT1 was evident. Finally, the CSF AEA level was related to CSF orexin levels in all subjects. CONCLUSION We demonstrated that the EC system is dysregulated in NT1.
Collapse
Affiliation(s)
- Andrea Romigi
- IRCCS Neuromed Sleep Medicine Center, Via Atinense, 18 Pozzilli (IS), Italy
| | - Monica Bari
- Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Francesca Izzi
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Cinzia Rapino
- Faculty of Bioscience and Technology for Food, University of Teramo, Agriculture and Environment, Teramo, Italy
| | - Marzia Nuccetelli
- Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Natalia Battista
- Faculty of Bioscience and Technology for Food, University of Teramo, Agriculture and Environment, Teramo, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Diego Centonze
- IRCCS Neuromed Sleep Medicine Center, Via Atinense, 18 Pozzilli (IS), Italy
| | | | - Fabio Placidi
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Mauro Maccarrone
- Campus Bio-Medico University of Rome, Italy.,European Center for Brain Research, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
39
|
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
|
40
|
Chen Q, Ji C, Zheng R, Yang L, Ren J, Li Y, Han Y, Zhou P, Liu Z, Qiu Y. N-Palmitoylethanolamine Maintains Local Lipid Homeostasis to Relieve Sleep Deprivation-Induced Dry Eye Syndrome. Front Pharmacol 2020; 10:1622. [PMID: 32047441 PMCID: PMC6997544 DOI: 10.3389/fphar.2019.01622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/13/2019] [Indexed: 11/13/2022] Open
Abstract
Sleep loss is a key factor associated with dry eye. Use of a “stick over water” mouse model revealed that sleep deprivation induces accumulation of lipids, hypertrophy, and dysfunction of the lacrimal gland. These changes result in decreased tear production and dry eye clinical signs. The specific pathophysiological mechanisms that contribute to dry eye remain unclear. In this study, we found that sleep deprivation decreased endogenous lipid palmitoylethanolamide (PEA) expression in the lacrimal gland. The reduced expression was mainly attributed to the decreased expression of N-acylated phosphatidylethanolamine–phospholipase D, the synthetic enzyme of PEA. Exogenous PEA treatment restored local lipid metabolism homeostasis in the lacrimal gland. This change was accompanied by reduced lipid deposition, maintenance of the endoplasmic reticulum and mitochondrial morphology, and improved acinar cell secretory function. PEA treatment also prevented damage to corneal barrier function and improved the dry eye clinical signs caused by sleep deprivation. The nuclear receptor peroxisome proliferator-activated receptor-α (PPAR-α) was found to mediate the PEA-associated improvements. We describe here for the first time that PEA is involved in sleep deprivation–induced lacrimal gland pathogenesis and dry eye development. PEA and its metabolizing enzymes may serve as adjunctive therapeutic targets for treatment of dry eye.
Collapse
Affiliation(s)
- Qi Chen
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Chunyan Ji
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Ruihe Zheng
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Longhe Yang
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen, China
| | - Jie Ren
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yitian Li
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yun Han
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Pan Zhou
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Zuguo Liu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Yan Qiu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| |
Collapse
|
41
|
Prospéro-García O, Ruiz Contreras AE, Ortega Gómez A, Herrera-Solís A, Méndez-Díaz M. Endocannabinoids as Therapeutic Targets. Arch Med Res 2020; 50:518-526. [PMID: 32028095 DOI: 10.1016/j.arcmed.2019.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/01/2019] [Accepted: 09/30/2019] [Indexed: 12/22/2022]
Abstract
Most of the drugs of abuse affect the brain by interacting with naturally expressed molecular receptors. Marihuana affects a series of receptors including cannabinoid receptor 1 (CB1R) and CB2R, among others. Endogenous molecules with cannabinoid activity interact with these receptors naturally. Receptors, ligands, synthesizing and degrading enzymes, as well as transporters, have been described. This endocannabinoid system modulates behaviors and physiological processes, i.e. food intake, the sleep-waking cycle, learning and memory, motivation, and pain perception, among others. The rather broad distribution of endocannabinoids in the brain explains the different effects marihuana induces in its users. However, this very same anatomical and physiological distribution makes this system a useful target for therapeutic endeavors. In this review, we briefly discuss the potential of small molecules that target the endocannabinoids as therapeutic tools to improve behaviors and treat illnesses. We believe that under medical supervision, endocannabinoid targets offer new advantages for patients for controlling multiple medical disorders.
Collapse
Affiliation(s)
- Oscar Prospéro-García
- Departamento de Fisiología, Laboratorio de Canabinoides, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - Alejandra E Ruiz Contreras
- Laboratorio de Neurogenómica Cognitiva, Facultad de Psicología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Alette Ortega Gómez
- Laboratorio de Medicina Traslacional, Instituto Nacional de Cancerología, Ciudad de México, Mexico
| | - Andrea Herrera-Solís
- Laboratorio Efectos Terapéuticos de los Canabinoides, Subdirección de Investigación Biomédica, Hospital General Dr. Manuel Gea González, Ciudad de México, Mexico
| | - Mónica Méndez-Díaz
- Departamento de Fisiología, Laboratorio de Canabinoides, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | | |
Collapse
|
42
|
Murillo-Rodríguez E, Budde H, Veras AB, Rocha NB, Telles-Correia D, Monteiro D, Cid L, Yamamoto T, Machado S, Torterolo P. The Endocannabinoid System May Modulate Sleep Disorders in Aging. Curr Neuropharmacol 2020; 18:97-108. [PMID: 31368874 PMCID: PMC7324886 DOI: 10.2174/1570159x17666190801155922] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
Aging is an inevitable process that involves changes across life in multiple neurochemical, neuroanatomical, hormonal systems, and many others. In addition, these biological modifications lead to an increase in age-related sickness such as cardiovascular diseases, osteoporosis, neurodegenerative disorders, and sleep disturbances, among others that affect activities of daily life. Demographic projections have demonstrated that aging will increase its worldwide rate in the coming years. The research on chronic diseases of the elderly is important to gain insights into this growing global burden. Novel therapeutic approaches aimed for treatment of age-related pathologies have included the endocannabinoid system as an effective tool since this biological system shows beneficial effects in preclinical models. However, and despite these advances, little has been addressed in the arena of the endocannabinoid system as an option for treating sleep disorders in aging since experimental evidence suggests that some elements of the endocannabinoid system modulate the sleep-wake cycle. This article addresses this less-studied field, focusing on the likely perspective of the implication of the endocannabinoid system in the regulation of sleep problems reported in the aged. We conclude that beneficial effects regarding the putative efficacy of the endocannabinoid system as therapeutic tools in aging is either inconclusive or still missing.
Collapse
Affiliation(s)
- Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mérida, Yucatán, México
- Intercontinental Neuroscience Research Group
| | - Henning Budde
- Intercontinental Neuroscience Research Group
- Faculty of Human Sciences, Medical School Hamburg, Hamburg, Germany
| | - André Barciela Veras
- Intercontinental Neuroscience Research Group
- Dom Bosco Catholic University, Campo Grande, Mato Grosso do Sul, Brazil
| | - Nuno Barbosa Rocha
- Intercontinental Neuroscience Research Group
- School of Health, Polytechnic Institute of Porto, Porto, Portugal
| | - Diogo Telles-Correia
- Intercontinental Neuroscience Research Group
- University of Lisbon, Faculty of Medicine, Lisbon, Portugal
| | - Diogo Monteiro
- Intercontinental Neuroscience Research Group
- Sport Science School of Rio Maior-Polytechnic Institute of Santarém, Rio Maior, Portugal
- Research Center in Sport, Health and Human Development-CIDESD, Vila Real, Portugal
| | - Luis Cid
- Intercontinental Neuroscience Research Group
- Sport Science School of Rio Maior-Polytechnic Institute of Santarém, Rio Maior, Portugal
- Research Center in Sport, Health and Human Development-CIDESD, Vila Real, Portugal
| | - Tetsuya Yamamoto
- Intercontinental Neuroscience Research Group
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Sérgio Machado
- Intercontinental Neuroscience Research Group
- Laboratory of Physical Activity Neuroscience, Physical Activity Sciences Postgraduate Program, Salgado de Oliveira University, Niterói, Brazil
| | - Pablo Torterolo
- Intercontinental Neuroscience Research Group
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
43
|
Hanlon EC. Impact of circadian rhythmicity and sleep restriction on circulating endocannabinoid (eCB) N-arachidonoylethanolamine (anandamide). Psychoneuroendocrinology 2020; 111:104471. [PMID: 31610409 PMCID: PMC7001881 DOI: 10.1016/j.psyneuen.2019.104471] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/26/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The endocannabinoid (eCB) system is involved in diverse aspects of human physiology and behavior but little is known about the impact of circadian rhythmicity on the system. The two most studied endocannabinoids, AEA (ananamide) and 2-AG (2-arachidonoylglycerol), can be measured in peripheral blood however the functional relevance of peripheral eCB levels is not clear. Having previously detailed the 24-h profile of serum 2-AG, here we report the 24-h serum profile of AEA to determine if these two endocannabinoids vary in parallel across the biological day including a nocturnal 8.5-h sleep period. Further, we assessed and compared the effect of a physiological challenge, in the form of sleep restriction to 4.5-h, on these two profiles. METHODS In this randomized crossover study, we examined serum concentrations of AEA across a 24-h period in fourteen young adults. Congeners of AEA, the structural analogs oleoylethanolamide (OEA) and palmitoylethanolamide (PEA) were simultaneously assayed. Prior to 24-h blood sampling, each participant was exposed to two nights of normal (8.5 h) or restricted sleep (4.5 h). The two sleep conditions were separated by at least one month. In both sleep conditions, during the period of blood sampling, each individual ate the same high-carbohydrate meal at 0900, 1400, and 1900. RESULTS Mean 24-h concentrations of AEA were 0.697 ± 0.11 pmol/ml. A reproducible biphasic 24-h profile of AEA was observed with a first peak occurring during early sleep (0200) and a second peak in the mid-afternoon (1500) while a nadir was detected in the mid-morning (1000). The 24-h profiles for both OEA and PEA followed a similar pattern to that observed for AEA. AEA, OEA, and PEA levels were not affected by sleep restriction at any time of day, contrasting with the elevation of early afternoon levels previously observed for 2-AG. CONCLUSIONS The 24-h rhythm of AEA is markedly different from that of 2-AG, being of lesser amplitude and biphasic, rather than monophasic. These observations suggest distinct regulatory pathways of the two eCB and indicate that time of day needs to be carefully controlled in studies attempting to delineate their relative roles. Moreover, unlike 2-AG, AEA is not altered by sleep restriction, suggesting that physiological perturbations may affect AEA and 2-AG differently. Similar 24-h profiles were observed for OEA and PEA following normal and restricted sleep, further corroborating the validity of the wave-shape and lack of response to sleep loss observed for the AEA profile. Therapeutic approaches involving agonism or antagonism of peripheral eCB signaling will likely need to be tailored according to time of day.
Collapse
Affiliation(s)
- Erin C Hanlon
- University of Chicago, Department of Medicine, MC 1027, Section of Endocrinology, Diabetes, and Metabolism, 5841 S Maryland Ave, Chicago, IL 60637, United States.
| |
Collapse
|
44
|
Nashed MG, Hardy DB, Laviolette SR. Prenatal Cannabinoid Exposure: Emerging Evidence of Physiological and Neuropsychiatric Abnormalities. Front Psychiatry 2020; 11:624275. [PMID: 33519564 PMCID: PMC7841012 DOI: 10.3389/fpsyt.2020.624275] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/14/2020] [Indexed: 12/22/2022] Open
Abstract
Clinical reports of cannabis use prevalence during pregnancy vary widely from 3% to upwards of 35% in North America; this disparity likely owing to underestimates from self-reporting in many cases. The rise in cannabis use is mirrored by increasing global legalization and the overall perceptions of safety, even during pregnancy. These trends are further compounded by a lack of evidence-based policy and guidelines for prenatal cannabis use, which has led to inconsistent messaging by healthcare providers and medically licensed cannabis dispensaries regarding prenatal cannabis use for treatment of symptoms, such as nausea. Additionally, the use of cannabis to self-medicate depression and anxiety during pregnancy is a growing medical concern. This review aims to summarize recent findings of clinical and preclinical data on neonatal outcomes, as well as long-term physiological and neurodevelopmental outcomes of prenatal cannabis exposure. Although many of the outcomes under investigation have produced mixed results, we consider these data in light of the unique challenges facing cannabis research. In particular, the limited longitudinal clinical studies available have not previously accounted for the exponential increase in (-)-Δ9- tetrahydrocannabinol (Δ9-THC; the psychoactive compound in cannabis) concentrations found in cannabis over the past two decades. Polydrug use and the long-term effects of individual cannabis constituents [Δ9-THC vs. cannabidiol (CBD)] are also understudied, along with sex-dependent outcomes. Despite these limitations, prenatal cannabis exposure has been linked to low birth weight, and emerging evidence suggests that prenatal exposure to Δ9-THC, which crosses the placenta and impacts placental development, may have wide-ranging physiological and neurodevelopmental consequences. The long-term effects of these changes require more rigorous investigation, though early reports suggest Δ9-THC increases the risk of cognitive impairment and neuropsychiatric disease, including psychosis, depression, anxiety, and sleep disorders. In light of the current trends in the perception and use of cannabis during pregnancy, we emphasize the social and medical imperative for more rigorous investigation of the long-term effects of prenatal cannabis exposure.
Collapse
Affiliation(s)
- Mina G Nashed
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Daniel B Hardy
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada.,Department of Obstetrics & Gynecology, University of Western Ontario, London, ON, Canada
| | - Steven R Laviolette
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada.,Department of Psychiatry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
45
|
Mondino A, Fernández S, Garcia-Carnelli C, Castro MJ, Umpierrez E, Torterolo P, Falconi A, Agrati D. Vaporized Cannabis differentially modulates sexual behavior of female rats according to the dose. Pharmacol Biochem Behav 2019; 187:172814. [PMID: 31644886 DOI: 10.1016/j.pbb.2019.172814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 09/01/2019] [Accepted: 10/15/2019] [Indexed: 10/25/2022]
Abstract
Studies exploring the effect of compounds that modulate the endocannabinoid system on sexual behavior have yielded contradictory results. However, the effect of smoked Cannabis in women has been consistently associated with an increase in sexual drive. Therefore, it can be speculated that vaporized Cannabis will augment sexually motivated components of the sexual behavior of female rats. To test this hypothesis, we compared the sexual behavior of late-proestrous female rats in a bilevel chamber after vaporizing 0, 200 or 400 mg of Cannabis flowers (containing 18% of delta-9-THC and undetectable levels of cannabidiol) during 10 min. We found that both doses of Cannabis increased the duration of the lordosis response, whereas the highest dose also reduced the lordosis quotient of females. The lowest dose of Cannabis augmented the display of hops and darts without altering the expression of sexual solicitations of females, while the highest one did not affect the expression of hops and darts but reduced sexual solicitations. These effects were not accompanied by alterations of females' ambulatory behavior. The increment of the duration of lordosis response produced by both doses of Cannabis could be associated to a general effect of this drug in sensory processing, as can be an enhancement of females' sensory reactivity to male's stimulation. However, the reduction in the display of solicitations and lordosis in response to mounting observed in females exposed to the highest dose when compared to control and 200 mg of Cannabis groups indicates a reduction of sexual receptivity and motivation. This differential effect of vaporized Cannabis according to the dose employed, suggests that it modulates sexual behavior in a complex way, impacting neural circuits that control different aspects of this social behavior.
Collapse
Affiliation(s)
- Alejandra Mondino
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, General Flores 2125, 11800 Montevideo, Uruguay.
| | - Santiago Fernández
- Laboratorio de Farmacognosia y Productos Naturales, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, General Flores 2124, 11800 Montevideo, Uruguay
| | - Carlos Garcia-Carnelli
- Laboratorio de Farmacognosia y Productos Naturales, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, General Flores 2124, 11800 Montevideo, Uruguay
| | - María José Castro
- Unidad de Medioambiente, Drogas y Doping, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Camino Aparicio Saravia S/N, Bypass ruta 8, 91000 Pando, Canelones, Uruguay
| | - Eleuterio Umpierrez
- Unidad de Medioambiente, Drogas y Doping, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República, Camino Aparicio Saravia S/N, Bypass ruta 8, 91000 Pando, Canelones, Uruguay
| | - Pablo Torterolo
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, General Flores 2125, 11800 Montevideo, Uruguay
| | - Atilio Falconi
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, General Flores 2125, 11800 Montevideo, Uruguay
| | - Daniella Agrati
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Iguá 4225, 11400 Montevideo, Uruguay.
| |
Collapse
|
46
|
Kleckner AS, Kleckner IR, Kamen CS, Tejani MA, Janelsins MC, Morrow GR, Peppone LJ. Opportunities for cannabis in supportive care in cancer. Ther Adv Med Oncol 2019; 11:1758835919866362. [PMID: 31413731 PMCID: PMC6676264 DOI: 10.1177/1758835919866362] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/03/2019] [Indexed: 12/17/2022] Open
Abstract
Cannabis has the potential to modulate some of the most common and debilitating symptoms of cancer and its treatments, including nausea and vomiting, loss of appetite, and pain. However, the dearth of scientific evidence for the effectiveness of cannabis in treating these symptoms in patients with cancer poses a challenge to clinicians in discussing this option with their patients. A review was performed using keywords related to cannabis and important symptoms of cancer and its treatments. Literature was qualitatively reviewed from preclinical models to clinical trials in the fields of cancer, human immunodeficiency virus (HIV), multiple sclerosis, inflammatory bowel disease, post-traumatic stress disorder (PTSD), and others, to prudently inform the use of cannabis in supportive and palliative care in cancer. There is a reasonable amount of evidence to consider cannabis for nausea and vomiting, loss of appetite, and pain as a supplement to first-line treatments. There is promising evidence to treat chemotherapy-induced peripheral neuropathy, gastrointestinal distress, and sleep disorders, but the literature is thus far too limited to recommend cannabis for these symptoms. Scant, yet more controversial, evidence exists in regard to cannabis for cancer- and treatment-related cognitive impairment, anxiety, depression, and fatigue. Adverse effects of cannabis are documented but tend to be mild. Cannabis has multifaceted potential bioactive benefits that appear to outweigh its risks in many situations. Further research is required to elucidate its mechanisms of action and efficacy and to optimize cannabis preparations and doses for specific populations affected by cancer.
Collapse
Affiliation(s)
- Amber S Kleckner
- Cancer Control and Survivorship, University of Rochester Medical Center, CU 420658, 265 Crittenden Blvd., Rochester, NY 14642, USA
| | - Ian R Kleckner
- Cancer Control and Survivorship, University of Rochester Medical Center, Rochester, NY, USA
| | - Charles S Kamen
- Cancer Control and Survivorship, University of Rochester Medical Center, Rochester, NY, USA
| | - Mohamedtaki A Tejani
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Michelle C Janelsins
- Cancer Control and Survivorship, University of Rochester Medical Center, Rochester, NY, USA
| | - Gary R Morrow
- Cancer Control and Survivorship, University of Rochester Medical Center, Rochester, NY, USA
| | - Luke J Peppone
- Cancer Control and Survivorship, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
47
|
Méndez-Díaz M, Amancio-Belmont O, Estrada-González V, Ruiz-Contreras AE, Prospéro-García O. CB1R mediates oleamide’s reward while 5HT2cR mediates aversion in the nucleus accumbens shell of rats. Neurosci Lett 2019; 706:189-193. [DOI: 10.1016/j.neulet.2019.05.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 10/26/2022]
|
48
|
Mondino A, Cavelli M, González J, Santana N, Castro-Zaballa S, Mechoso B, Bracesco N, Fernandez S, Garcia-Carnelli C, Castro MJ, Umpierrez E, Murillo-Rodriguez E, Torterolo P, Falconi A. Acute effect of vaporized Cannabis on sleep and electrocortical activity. Pharmacol Biochem Behav 2019; 179:113-123. [DOI: 10.1016/j.pbb.2019.02.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/04/2019] [Accepted: 02/24/2019] [Indexed: 01/31/2023]
|
49
|
Bogáthy E, Papp N, Tóthfalusi L, Vas S, Bagdy G. Additive effect of 5-HT2C and CB1 receptor blockade on the regulation of sleep-wake cycle. BMC Neurosci 2019; 20:14. [PMID: 30894126 PMCID: PMC6427841 DOI: 10.1186/s12868-019-0495-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/12/2019] [Indexed: 12/22/2022] Open
Abstract
Background Previous data show that serotonin 2C (5-HT2C) and cannabinoid 1 (CB1) receptors have a role in the modulation of sleep–wake cycle. Namely, antagonists on these receptors promoted wakefulness and inhibited rapid eye movement sleep (REMS) in rodents. The interaction of these receptors are also present in other physiological functions, such as the regulation of appetite. Blockade of 5-HT2C receptors modulat the effect of CB1 receptor antagonist, presumably in consecutive or interdependent steps. Here we investigate, whether previous blockade of 5-HT2C receptors can affect CB1 receptor functions in the sleep–wake regulation. Results Wistar rats were equipped with electroencephalography (EEG) and electromyography (EMG) electrodes. Following the recovery and habituation after surgery, animals were injected intraperitoneally (ip.) with SB-242084, a 5-HT2C receptor antagonist (1.0 mg/kg) at light onset (beginning of passive phase) followed by an injection with AM-251, a CB1 receptor antagonist (5.0 or 10.0 mg/kg, ip.) 10 min later. EEG, EMG and motor activity were analyzed for the subsequent 2 h. Both SB-242084 and AM-251 increased the time spent in active wakefulness, while decreased the time spent in non-REMS and REMS stages in the first 2 h of passive phase. In combination, the effect of the agents were additive, furthermore, statistical analysis did not show any interaction between the effects of these drugs in the modulation of vigilance stages. Conclusions Our results suggest that 5-HT2C receptor blockade followed by blockade of CB1 receptors evoked additive effect on the regulation of sleep–wake pattern.
Collapse
Affiliation(s)
- Emese Bogáthy
- Department of Pharmacodynamics, Semmelweis University, Budapest, Nagyvárad tér 4, 1089, Hungary
| | - Noémi Papp
- Department of Pharmacodynamics, Semmelweis University, Budapest, Nagyvárad tér 4, 1089, Hungary
| | - Laszló Tóthfalusi
- Department of Pharmacodynamics, Semmelweis University, Budapest, Nagyvárad tér 4, 1089, Hungary
| | - Szilvia Vas
- Department of Pharmacodynamics, Semmelweis University, Budapest, Nagyvárad tér 4, 1089, Hungary.,MTA-SE, Neuropsychopharmacology and Neurochemistry Research Group, Budapest, 1089, Hungary.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
| | - György Bagdy
- Department of Pharmacodynamics, Semmelweis University, Budapest, Nagyvárad tér 4, 1089, Hungary. .,MTA-SE, Neuropsychopharmacology and Neurochemistry Research Group, Budapest, 1089, Hungary. .,NAP-A-SE, New Antidepressant Target Research Group, Budapest, 1089, Hungary. .,NAP-2-SE, New Antidepressant Target Research Group, Budapest, 1089, Hungary.
| |
Collapse
|
50
|
Schonhofen P, Bristot IJ, Crippa JA, Hallak JEC, Zuardi AW, Parsons RB, Klamt F. Cannabinoid-Based Therapies and Brain Development: Potential Harmful Effect of Early Modulation of the Endocannabinoid System. CNS Drugs 2018; 32:697-712. [PMID: 30109642 DOI: 10.1007/s40263-018-0550-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The endocannabinoid retrograde signaling pathway is widely expressed in the central nervous system, where it plays major roles in regulating synaptic plasticity (excitatory and inhibitory) through long-term potentiation and long-term depression. The endocannabinoid system (ECS) components-cannabinoid receptors, endocannabinoids and synthesis/degradation enzymes-are expressed and are functional from early developmental stages and throughout adolescent cortical development, regulating progenitor cell fate, neural differentiation, migration and survival. This may potentially confer increased vulnerability to adverse outcomes from early cannabinoid exposure. Cannabidiol (CBD) is one of the most studied exogenous cannabinoids, and CBD-enriched Cannabis extracts have been widely (and successfully) used as adjuvants to treat children with refractory epilepsy, and there is even a Food and Drug Administration (FDA)-approved drug with purified CBD derived from Cannabis. However, there is insufficient information on possible long-term changes in the central nervous system caused by cannabinoid treatments during early childhood. Like the majority of cannabinoids, CBD is able to exert its effects directly and indirectly through the ECS, which can perturb the regulatory processes mediated by this system. In addition, CBD has a large number of non-endocannabinoid targets, which can explain CBD's effects. Here, we review the current knowledge about CBD-based therapies-pure and CBD-enriched Cannabis extracts-in studies with pediatric patients, their side effects, and their mechanisms of action regarding the central nervous system and neurodevelopment aspects. Since Cannabis extracts contain Δ9-tetrahydrocannabinol (Δ9-THC), we consider that pure CBD is possibly safer for young patients. Nevertheless, CBD, as well as other natural and/or synthetic cannabinoids, should be studied in more detail as a therapeutic alternative to CBD-enriched Cannabis extracts for young patients.
Collapse
Affiliation(s)
- Patrícia Schonhofen
- Laboratory of Cellular Biochemistry, Department of Biochemistry, ICBS/UFRGS, 2600 Ramiro Barcelos St, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, ICBS/UFRGS, Porto Alegre, RS, 90035-003, Brazil
- National Institutes of Science and Technology-Translational Medicine (INCT-TM), Porto Alegre, Brazil
| | - Ivi Juliana Bristot
- Laboratory of Cellular Biochemistry, Department of Biochemistry, ICBS/UFRGS, 2600 Ramiro Barcelos St, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, ICBS/UFRGS, Porto Alegre, RS, 90035-003, Brazil
- National Institutes of Science and Technology-Translational Medicine (INCT-TM), Porto Alegre, Brazil
| | - José Alexandre Crippa
- National Institutes of Science and Technology-Translational Medicine (INCT-TM), Porto Alegre, Brazil
- Neuroscience and Behavior Department, Faculty of Medicine of Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Jaime Eduardo Cecílio Hallak
- National Institutes of Science and Technology-Translational Medicine (INCT-TM), Porto Alegre, Brazil
- Neuroscience and Behavior Department, Faculty of Medicine of Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Antônio Waldo Zuardi
- National Institutes of Science and Technology-Translational Medicine (INCT-TM), Porto Alegre, Brazil
- Neuroscience and Behavior Department, Faculty of Medicine of Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Richard B Parsons
- Institute of Pharmaceutical Science, King's College London (KCL), London, SE1 9NH, UK
| | - Fábio Klamt
- Laboratory of Cellular Biochemistry, Department of Biochemistry, ICBS/UFRGS, 2600 Ramiro Barcelos St, Porto Alegre, RS, 90035-003, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, ICBS/UFRGS, Porto Alegre, RS, 90035-003, Brazil.
- National Institutes of Science and Technology-Translational Medicine (INCT-TM), Porto Alegre, Brazil.
| |
Collapse
|