1
|
Gamble MC, Miracle S, Williams BR, Logan RW. Endocannabinoid agonist 2-arachidonoylglycerol differentially alters diurnal activity and sleep during fentanyl withdrawal in male and female mice. Pharmacol Biochem Behav 2024; 240:173791. [PMID: 38761993 PMCID: PMC11166043 DOI: 10.1016/j.pbb.2024.173791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Fentanyl has become the leading driver of opioid overdoses in the United States. Cessation of opioid use represents a challenge as the experience of withdrawal drives subsequent relapse. One of the most prominent withdrawal symptoms that can contribute to opioid craving and vulnerability to relapse is sleep disruption. The endocannabinoid agonist, 2-Arachidonoylglycerol (2-AG), may promote sleep and reduce withdrawal severity; however, the effects of 2-AG on sleep disruption during opioid withdrawal have yet to be assessed. Here, we investigated the effects of 2-AG administration on sleep-wake behavior and diurnal activity in mice during withdrawal from fentanyl. Sleep-wake activity measured via actigraphy was continuously recorded before and after chronic fentanyl administration in both male and female C57BL/6J mice. Immediately following cessation of fentanyl administration, 2-AG was administered intraperitoneally to investigate the impact of endocannabinoid agonism on opioid-induced sleep disruption. We found that female mice maintained higher activity levels in response to chronic fentanyl than male mice. Furthermore, fentanyl administration increased wake and decreased sleep during the light period and inversely increased sleep and decreased wake in the dark period in both sexes. 2-AG treatment increased arousal and decreased sleep in both sexes during first 24-h of withdrawal. On withdrawal day 2, only females showed increased wakefulness with no changes in males, but by withdrawal day 3 male mice displayed decreased rapid-eye movement sleep during the dark period with no changes in female mice. Overall, repeated administration of fentanyl altered sleep and diurnal activity and administration of the endocannabinoid agonist, 2-AG, had sex-specific effects on fentanyl-induced sleep and diurnal changes.
Collapse
Affiliation(s)
- Mackenzie C Gamble
- Molecular and Translational Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Sophia Miracle
- Graduate Program in Neuroscience, Boston University, Boston, MA, USA
| | - Benjamin R Williams
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ryan W Logan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
2
|
Hebert FO, Mongeau-Pérusse V, Rizkallah E, Mahroug A, Bakouni H, Morissette F, Brissette S, Bruneau J, Dubreucq S, Jutras-Aswad D. Absence of Evidence for Sustained Effects of Daily Cannabidiol Administration on Anandamide Plasma Concentration in Individuals with Cocaine Use Disorder: Exploratory Findings from a Randomized Controlled Trial. Cannabis Cannabinoid Res 2024. [PMID: 38770686 DOI: 10.1089/can.2023.0273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
Background: Cannabidiol (CBD) has been proposed to have a therapeutic potential over a wide range of neuropsychiatric disorders, including substance use disorders. Pre-clinical evidence suggests that CBD can increase anandamide (AEA) plasma concentration, possibly mediating some of its therapeutic properties. Whether CBD exerts such an effect on AEA in individuals with cocaine use disorder (CUD) remains unknown. Aims: To explore the sustained effects of daily CBD administration on AEA plasma concentrations compared with placebo in CUD. Methods: We used data from a randomized, double-blind, placebo-controlled trial evaluating CBD's efficacy in CUD. Seventy-eight individuals were randomized to receive a daily oral dose of 800 mg CBD (n = 40) or a placebo (n = 38). Participants stayed in an inpatient detoxification setting for 10 days, after which they were followed in an outpatient setting for 12 weeks. AEA plasma concentration was measured at baseline and at 23-h post CBD ingestion on day 8 and week 4. A generalized estimating equation model was used to assess CBD's effects on AEA, and sensitivity analyses were computed using Bayesian linear regressions. Results: Sixty-four participants were included in the analysis. Similar mean AEA plasma concentrations in both treatment groups (p = 0.357) were observed. At day 8, mean AEA plasma concentrations (± standard deviation) were 0.26 (± 0.07) ng/mL in the CBD group and 0.29 (± 0.08) ng/mL in the placebo group (p = 0.832; Bayes factor [BF] = 0.190). At week 4, they were 0.27 (± 0.09) ng/mL in the CBD group and 0.30 (± 0.09) ng/mL in the placebo group (p = 0.181; BF = 0.194). Conclusion: While not excluding any potential acute and short-term effect, daily CBD administration did not exert a sustained impact on AEA plasma concentrations in individuals with CUD compared with placebo. Registration: clinicaltrials.gov (NCT02559167).
Collapse
Affiliation(s)
| | - Violaine Mongeau-Pérusse
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Elie Rizkallah
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Amani Mahroug
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
| | - Hamzah Bakouni
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Florence Morissette
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Suzanne Brissette
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Family and Emergency Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Julie Bruneau
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Family and Emergency Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Simon Dubreucq
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Didier Jutras-Aswad
- Research Center, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Department of Psychiatry and Addiction, Faculty of Medicine, Université de Montréal, Montréal, Canada
| |
Collapse
|
3
|
Scicluna RL, Wilson BB, Thelaus SH, Arnold JC, McGregor IS, Bowen MT. Cannabidiol Reduced the Severity of Gastrointestinal Symptoms of Opioid Withdrawal in Male and Female Mice. Cannabis Cannabinoid Res 2024; 9:547-560. [PMID: 36577048 DOI: 10.1089/can.2022.0036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Introduction: Opioid withdrawal is a powerful driver of drug-seeking behavior as relief from this aversive state through drug-taking is a strong negative reinforcer. There are currently limited treatment options available for opioid withdrawal and cannabidiol (CBD) has been identified as a potential novel therapeutic. This study explored the efficacy and dose dependency of CBD for reducing the severity of naloxone-precipitated and spontaneous oxycodone withdrawal (PW and SW, respectively) in male and female mice. Methods: Mice were administered saline or escalating doses of oxycodone, whereby 9, 17.8, 23.7, and 33 mg/kg oxycodone IP was administered twice daily on days 1-2, 3-4, 5-6, and 7-8, respectively. On the 9th day, a single 33 mg/kg dose of oxycodone (or saline) was administered. To precipitate withdrawal, on day 9, mice in the withdrawal conditions were administered an IP injection of 10 mg/kg naloxone 2 h after the final oxycodone injection and immediately before withdrawal testing. To elicit SW, a separate group of mice underwent withdrawal testing 24 h after their final oxycodone injection. Mice were treated with an IP injection of 0, 10, 30 or 100 mg/kg of CBD 60 min before testing. Withdrawal symptoms examined included gastrointestinal symptoms (fecal boli, diarrhea, and body weight loss), somatic symptoms (paw tremors), and negative affect (jumping). Results: A robust PW syndrome was observed in both male and female mice, whereas only male mice displayed an SW syndrome. CBD dose dependently reduced gastrointestinal symptoms during both PW and SW in male mice and during PW in female mice. CBD had no effect on PW- or SW-induced jumping in male mice. However, in female mice, the PW-induced increase in jumps was less pronounced in CBD-treated mice. The highest dose of CBD inhibited paw tremors during PW, but not SW, in male mice. Neither PW- nor SW-induced paw tremors were observed in female mice. Conclusions: The magnitude of effects on the gastrointestinal symptoms, their consistency across PW and SW, and both sexes, alongside the availability of CBD for clinical use, suggest further exploration of the potential for CBD to treat these symptoms could be justified.
Collapse
Affiliation(s)
- Rhianne L Scicluna
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Bianca B Wilson
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Samuel H Thelaus
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Jonathon C Arnold
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Pharmacy, Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, New South Wales, Australia
| | - Iain S McGregor
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, New South Wales, Australia
| | - Michael T Bowen
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Pota V, Sansone P, De Sarno S, Aurilio C, Coppolino F, Barbarisi M, Barbato F, Fiore M, Cosenza G, Passavanti MB, Pace MC. Amyotrophic Lateral Sclerosis and Pain: A Narrative Review from Pain Assessment to Therapy. Behav Neurol 2024; 2024:1228194. [PMID: 38524401 PMCID: PMC10960655 DOI: 10.1155/2024/1228194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 02/11/2024] [Accepted: 03/06/2024] [Indexed: 03/26/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most frequent neurodegenerative disease of the motor system that affects upper and lower motor neurons, leading to progressive muscle weakness, spasticity, atrophy, and respiratory failure, with a life expectancy of 2-5 years after symptom onset. In addition to motor symptoms, patients with ALS have a multitude of nonmotor symptoms; in fact, it is currently considered a multisystem disease. The purpose of our narrative review is to evaluate the different types of pain, the correlation between pain and the disease's stages, the pain assessment tools in ALS patients, and the available therapies focusing above all on the benefits of cannabis use. Pain is an underestimated and undertreated symptom that, in the last few years, has received more attention from research because it has a strong impact on the quality of life of these patients. The prevalence of pain is between 15% and 85% of ALS patients, and the studies on the type and intensity of pain are controversial. The absence of pain assessment tools validated in the ALS population and the dissimilar study designs influence the knowledge of ALS pain and consequently the pharmacological therapy. Several studies suggest that ALS is associated with changes in the endocannabinoid system, and the use of cannabis could slow the disease progression due to its neuroprotective action and act on pain, spasticity, cramps, sialorrhea, and depression. Our research has shown high patients' satisfaction with the use of cannabis for the treatment of spasticity and related pain. However, especially due to the ethical problems and the lack of interest of pharmaceutical companies, further studies are needed to ensure the most appropriate care for ALS patients.
Collapse
Affiliation(s)
- Vincenzo Pota
- Department of Women, Child, General and Specialistic Surgery, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Pasquale Sansone
- Department of Women, Child, General and Specialistic Surgery, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Sara De Sarno
- Department of Women, Child, General and Specialistic Surgery, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Caterina Aurilio
- Department of Women, Child, General and Specialistic Surgery, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Francesco Coppolino
- Department of Women, Child, General and Specialistic Surgery, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Manlio Barbarisi
- Multidisciplinary Department of Medical, Surgical and Dental Specialties, University of Campania “L. Vanvitelli”, Naples, Italy
| | | | - Marco Fiore
- Department of Women, Child, General and Specialistic Surgery, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Gianluigi Cosenza
- Department of Women, Child, General and Specialistic Surgery, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Maria Beatrice Passavanti
- Department of Women, Child, General and Specialistic Surgery, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Maria Caterina Pace
- Department of Women, Child, General and Specialistic Surgery, University of Campania “L. Vanvitelli”, Naples, Italy
| |
Collapse
|
5
|
Gamble MC, Miracle S, Williams BR, Logan RW. Sex-specific Effects of the Endocannabinoid Agonist 2-Arachidonoylglycerol on Sleep and Circadian Disruptions during Fentanyl Withdrawal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.19.572466. [PMID: 38187736 PMCID: PMC10769247 DOI: 10.1101/2023.12.19.572466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Fentanyl has become the leading driver of opioid overdoses. Cessation of opioid use represents a challenge as the experience of withdrawal drives subsequent relapse. One of the most prominent withdrawal symptoms that can contribute to opioid craving and vulnerability to relapse is sleep disruption. The endocannabinoid agonist, 2-Arachidonoylglycerol (2-AG), may promote sleep and reduce withdrawal severity; however, the effects of 2-AG on sleep disruption during opioid withdrawal have yet to be assessed. Here, we investigate the effects of 2-AG administration on sleep-wake behavior and diurnal activity in mice during withdrawal from fentanyl. Sleep-wake activity was continuously recorded before and after chronic fentanyl administration in both male and female C57BL/6J mice. Immediately following cessation of fentanyl administration, 2-AG was administered intraperitoneally to investigate the impact of endocannabinoid agonism on opioid-induced sleep disruption. Female mice maintained higher activity levels in response to chronic fentanyl than male mice. Furthermore, fentanyl increased wake and decreased sleep during the light period and inversely increased sleep and decreased wake in the dark period in both sexes. 2-AG treatment increased arousal and decreased sleep in both sexes during first 24 hrs of withdrawal. On withdrawal day 2, only female showed increased wakefulness with no changes in males, but by withdrawal day 3 male mice displayed decreased rapid-eye movement sleep during the dark period with no changes in female mice. Overall, repeated administration of fentanyl altered sleep and diurnal activity and administration of the endocannabinoid agonist, 2-AG, had sex-specific effects on fentanyl-induced sleep and diurnal changes.
Collapse
|
6
|
Jaiswal S, Akhilesh, Tiwari V, Ayyannan SR. Anti-nociceptive potential of an isatin-derived dual fatty acid amide hydrolase-monoacylglycerol lipase inhibitor. Pharmacol Rep 2023; 75:737-745. [PMID: 36913176 DOI: 10.1007/s43440-023-00468-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/14/2023]
Abstract
BACKGROUND Recently, we have reported an isatin-derived carbohydrazone, 5-chloro-N'-(6-chloro-2-oxoindolin-3-ylidene)-2-hydroxybenzohydrazide (SIH 3) as dual nanomolar FAAH (fatty acid amide hydrolase)-MAGL (monoacylglycerol lipase) inhibitor with good CNS penetration and neuroprotective activity profile. In this study, we further investigated the pharmacological profile of compound SIH 3 in the neuropathic pain model along with acute toxicity and ex vivo studies. METHODS Chronic constrictive injury (CCI) was used to induce neuropathic pain in male Sprague-Dawley rats and the anti-nociceptive activity of the compound SIH 3 was investigated at 25, 50, and 100 mg/kg ip. Subsequently, locomotor activity was measured by rotarod and actophotometer experiments. The acute oral toxicity of the compound was assessed as per the OECD guidelines 423. RESULTS Compound SIH 3 showed significant anti-nociceptive activity in the CCI-induced neuropathic pain model without altering the locomotor activity. Furthermore, compound SIH 3 showed an excellent safety profile (up to 2000 mg/kg, po) in the acute oral toxicity study and was also non-hepatotoxic. Further, ex vivo studies revealed that the compound SIH 3 produces a significant antioxidant effect in oxidative stress induced by CCI. CONCLUSION Our findings suggest that the investigated compound SIH 3 has the potential to be developed as an anti-nociceptive agent.
Collapse
Affiliation(s)
- Shivani Jaiswal
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, UP, 221005, India
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, 281406, India
| | - Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, UP, 221005, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, UP, 221005, India
| | - Senthil Raja Ayyannan
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, UP, 221005, India.
| |
Collapse
|
7
|
de Ceglia M, Micioni Di Bonaventura MV, Romano A, Friuli M, Micioni Di Bonaventura E, Gavito AL, Botticelli L, Gaetani S, de Fonseca FR, Cifani C. Anxiety associated with palatable food withdrawal is reversed by the selective FAAH inhibitor PF-3845: A regional analysis of the contribution of endocannabinoid signaling machinery. Int J Eat Disord 2023. [PMID: 36840536 DOI: 10.1002/eat.23917] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/26/2023]
Abstract
OBJECTIVE Consumption of energy-dense palatable "comfort" food can alleviate stress and negative emotions, while abrupt withdrawal from a palatable diet can worsen these symptoms, causing difficulties with adherence to weight-loss diets. Currently, no pharmacological treatment is effective for obesity-related anxiety, so we investigated the endocannabinoid system (ECS), and specifically the fatty acid amide hydrolase (FAAH), as an interesting emerging target in this context because of its key role in the regulation of both energy homeostasis and emotional behavior. METHODS Rats were subjected to exposure and subsequent abstinence from a palatable cafeteria diet. During abstinence period, rats were treated with the selective FAAH inhibitor PF-3845 (10 mg/kg; intraperitoneal administration every other day). RESULTS Abstinent rats displayed an anxiogenic-like behavior and changes in the proteins of ECS signaling machinery in brain areas involved both in anxiety and food intake regulation. In particular, withdrawal caused a reduction of the expression of cannabinoid receptors in the nucleus accumbens and of enzymes diacylglycerol lipase alpha and monoacylglycerol lipase (MAGL) in the amygdala. Pharmacological inhibition of FAAH exerted an anxiolytic-like effect in abstinent animals and increased both MAGL expression in amygdala and CB2 expression in prefrontal cortex. DISCUSSION Overall, our results suggest that emotional disturbances associated with dieting are coupled with region-specific alterations in the cerebral expression of the ECS and that the enhancement of the endocannabinoid signaling by FAAH inhibition might represent a novel pharmacological strategy for the treatment of anxiety related to abstinence from palatable food. PUBLIC SIGNIFICANCE The present study focused on evaluating the role of the endocannabinoid system in modulating withdrawal from naturally rewarding activities that have an impact on mood, such as feeding. The variations observed in the emotional behavior of abstinent rats was linked to neuroadaptations of the ECS in specific brain areas.
Collapse
Affiliation(s)
- Marialuisa de Ceglia
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga-Hospital Universitario Regional de Málaga, Málaga, Spain.,Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | | | - Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Marzia Friuli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | | | - Ana L Gavito
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga-Hospital Universitario Regional de Málaga, Málaga, Spain
| | - Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga-Hospital Universitario Regional de Málaga, Málaga, Spain
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| |
Collapse
|
8
|
Jaiswal S, Gupta G, Ayyannan SR. Synthesis and evaluation of carbamate derivatives as fatty acid amide hydrolase and monoacylglycerol lipase inhibitors. Arch Pharm (Weinheim) 2022; 355:e2200081. [PMID: 35924298 DOI: 10.1002/ardp.202200081] [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: 02/14/2022] [Revised: 06/24/2022] [Accepted: 07/11/2022] [Indexed: 11/06/2022]
Abstract
Fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) are the primary catabolic enzymes for endocannabinoids, anandamide (AEA), and 2-arachidonoyl glycerol. Numerous studies have shown that FAAH and MAGL play an important role in modulating various central nervous system activities; hence, the development of small molecule FAAH/MAGL inhibitors is an active area of research. Several small molecules possessing the carbamate scaffold are documented as potential FAAH/MAGL inhibitors. Here, we designed and synthesized a series of open chain and cyclic carbamates and evaluated their dual FAAH-MAGL inhibition properties. Phenyl [4-(piperidin-1-ylmethyl)phenyl]carbamate (2e) emerged as the most potent MAGL inhibitor (IC50 = 19 nM), benzyl (1H-benzo[d]imidazol-2-yl)carbamate (3h) was the most potent FAAH inhibitor (IC50 = 55 nM), and phenyl (6-fluorobenzo[d]thiazol-2-yl)carbamate (2i) egressed as a nonselective dual FAAH-MAGL inhibitor (FAAH: 82 nM, MAGL: 72 nM). The enzyme kinetics experiments revealed that the compounds inhibit FAAH/MAGL in a covalent-reversible manner, with a mixed binding mode of action. Moreover, the lead compounds were found suitable for blood-brain permeation in the parallel artificial membrane permeation assay. Furthermore, docking simulation experiments suggested that the potency of the lead compounds was governed by hydrogen bonds and hydrophobic interactions with the enzyme active sites. In silico drug-likeness and ADMETox prediction studies provided useful information on the compounds' oral absorption, metabolism, and toxicity profiles. In summary, this study afforded potent multifunctional carbamates with appreciable pharmacokinetic profiles meriting further investigation.
Collapse
Affiliation(s)
- Shivani Jaiswal
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Garima Gupta
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Senthil R Ayyannan
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| |
Collapse
|
9
|
Papa A, Pasquini S, Contri C, Gemma S, Campiani G, Butini S, Varani K, Vincenzi F. Polypharmacological Approaches for CNS Diseases: Focus on Endocannabinoid Degradation Inhibition. Cells 2022; 11:cells11030471. [PMID: 35159280 PMCID: PMC8834510 DOI: 10.3390/cells11030471] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 01/27/2023] Open
Abstract
Polypharmacology breaks up the classical paradigm of “one-drug, one target, one disease” electing multitarget compounds as potential therapeutic tools suitable for the treatment of complex diseases, such as metabolic syndrome, psychiatric or degenerative central nervous system (CNS) disorders, and cancer. These diseases often require a combination therapy which may result in positive but also negative synergistic effects. The endocannabinoid system (ECS) is emerging as a particularly attractive therapeutic target in CNS disorders and neurodegenerative diseases including Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), stroke, traumatic brain injury (TBI), pain, and epilepsy. ECS is an organized neuromodulatory network, composed by endogenous cannabinoids, cannabinoid receptors type 1 and type 2 (CB1 and CB2), and the main catabolic enzymes involved in the endocannabinoid inactivation such as fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL). The multiple connections of the ECS with other signaling pathways in the CNS allows the consideration of the ECS as an optimal source of inspiration in the development of innovative polypharmacological compounds. In this review, we focused our attention on the reported polypharmacological examples in which FAAH and MAGL inhibitors are involved.
Collapse
Affiliation(s)
- Alessandro Papa
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
| | - Silvia Pasquini
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| | - Chiara Contri
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
- Correspondence: ; Tel.: +39-0577-234161
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| |
Collapse
|
10
|
Karimi-Haghighi S, Razavi Y, Iezzi D, Scheyer AF, Manzoni O, Haghparast A. Cannabidiol and substance use disorder: Dream or reality. Neuropharmacology 2022; 207:108948. [PMID: 35032495 PMCID: PMC9157244 DOI: 10.1016/j.neuropharm.2022.108948] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Cannabidiol (CBD) is one of the major constituents of Cannabis sativa L. that lacks psychotomimetic and rewarding properties and inhibits the rewarding and reinforcing effects of addictive drugs such as cocaine, methamphetamine (METH), and morphine. Additionally, CBD's safety profile and therapeutic potential are currently evaluated in several medical conditions, including pain, depression, movement disorders, epilepsy, multiple sclerosis, Alzheimer's disease, ischemia, and substance use disorder. There is no effective treatment for substance use disorders such as addiction, and this review aims to describe preclinical and clinical investigations into the effects of CBD in various models of opioid, psychostimulant, cannabis, alcohol, and nicotine abuse. Furthermore, the possible mechanisms underlying the therapeutic potential of CBD on drug abuse disorders are reviewed. METHODS The current review considers and summarizes the preclinical and clinical investigations into CBD's effects in various models of drug abuse include opioids, psychostimulants, cannabis, alcohol, and nicotine. RESULTS Several preclinical and clinical studies have proposed that CBD may be a reliable agent to inhibit the reinforcing and rewarding impact of drugs. CONCLUSIONS While the currently available evidence converges to suggest that CBD could effectively reduce the rewarding and reinforcing effects of addictive drugs, more preclinical and clinical studies are needed before CBD can be added to the therapeutic arsenal for treating addiction.
Collapse
Affiliation(s)
- Saeideh Karimi-Haghighi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yasaman Razavi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Daniela Iezzi
- INMED, INSERM U1249, Marseille, France; Aix-Marseille University, Marseille, France
| | - Andrew F Scheyer
- INMED, INSERM U1249, Marseille, France; Aix-Marseille University, Marseille, France
| | - Olivier Manzoni
- INMED, INSERM U1249, Marseille, France; Aix-Marseille University, Marseille, France
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Dodu JC, Moncayo RK, Damaj MI, Schlosburg JE, Akbarali HI, O'Brien LD, Kendall DA, Wu Z, Lu D, Lichtman AH. The Cannabinoid Receptor Type 1 Positive Allosteric Modulator ZCZ011 Attenuates Naloxone-Precipitated Diarrhea and Weight Loss in Oxycodone-Dependent Mice. J Pharmacol Exp Ther 2022; 380:1-14. [PMID: 34625464 PMCID: PMC8969135 DOI: 10.1124/jpet.121.000723] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/01/2021] [Indexed: 01/03/2023] Open
Abstract
Opioid use disorder reflects a major public health crisis of morbidity and mortality in which opioid withdrawal often contributes to continued use. However, current medications that treat opioid withdrawal symptoms are limited by their abuse liability or lack of efficacy. Although cannabinoid 1 (CB1) receptor agonists, including Δ9-tetrahydrocannabinol, ameliorate opioid withdrawal in both clinical and preclinical studies of opioid dependence, this strategy elicits cannabimimetic side effects as well as tolerance and dependence after repeated administration. Alternatively, CB1 receptor positive allosteric modulators (PAMs) enhance CB1 receptor signaling and show efficacy in rodent models of pain and cannabinoid dependence but lack cannabimimetic side effects. We hypothesize that the CB1 receptor PAM ZCZ011 attenuates naloxone-precipitated withdrawal signs in opioid-dependent mice. Accordingly, male and female mice given an escalating dosing regimen of oxycodone, a widely prescribed opioid, and challenged with naloxone displayed withdrawal signs that included diarrhea, weight loss, jumping, paw flutters, and head shakes. ZCZ011 fully attenuated naloxone-precipitated withdrawal-induced diarrhea and weight loss and reduced paw flutters by approximately half, but its effects on head shakes were unreliable, and it did not affect jumping behavior. The antidiarrheal and anti-weight loss effects of ZCZ0111 were reversed by a CB1 not a cannabinoid receptor type 2 receptor antagonist and were absent in CB1 (-/-) mice, suggesting a necessary role of CB1 receptors. Collectively, these results indicate that ZCZ011 completely blocked naloxone-precipitated diarrhea and weight loss in oxycodone-dependent mice and suggest that CB1 receptor PAMs may offer a novel strategy to treat opioid dependence. SIGNIFICANCE STATEMENT: Opioid use disorder represents a serious public health crisis in which current medications used to treat withdrawal symptoms are limited by abuse liability and side effects. The CB1 receptor positive allosteric modulator (PAM) ZCZ011, which lacks overt cannabimimetic behavioral effects, ameliorated naloxone-precipitated withdrawal signs through a CB1 receptor mechanism of action in a mouse model of oxycodone dependence. These results suggest that CB1 receptor PAMs may represent a viable strategy to treat opioid withdrawal.
Collapse
Affiliation(s)
- Julien C Dodu
- Department of Pharmacology & Toxicology (J.C.D., R.K.M., M.I.D., J.E.S., H.I.A., L.D.O., A.H.L.), and Department of Medicinal Chemistry (A.H.L.), Virginia Commonwealth University, Richmond, Virginia; Department of Pharmaceutical Sciences, University of Connecticut, Mansfield, Connecticut (D.A.K.); and Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas (Z.W., D.L.)
| | - Rebecca K Moncayo
- Department of Pharmacology & Toxicology (J.C.D., R.K.M., M.I.D., J.E.S., H.I.A., L.D.O., A.H.L.), and Department of Medicinal Chemistry (A.H.L.), Virginia Commonwealth University, Richmond, Virginia; Department of Pharmaceutical Sciences, University of Connecticut, Mansfield, Connecticut (D.A.K.); and Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas (Z.W., D.L.)
| | - M Imad Damaj
- Department of Pharmacology & Toxicology (J.C.D., R.K.M., M.I.D., J.E.S., H.I.A., L.D.O., A.H.L.), and Department of Medicinal Chemistry (A.H.L.), Virginia Commonwealth University, Richmond, Virginia; Department of Pharmaceutical Sciences, University of Connecticut, Mansfield, Connecticut (D.A.K.); and Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas (Z.W., D.L.)
| | - Joel E Schlosburg
- Department of Pharmacology & Toxicology (J.C.D., R.K.M., M.I.D., J.E.S., H.I.A., L.D.O., A.H.L.), and Department of Medicinal Chemistry (A.H.L.), Virginia Commonwealth University, Richmond, Virginia; Department of Pharmaceutical Sciences, University of Connecticut, Mansfield, Connecticut (D.A.K.); and Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas (Z.W., D.L.)
| | - Hamid I Akbarali
- Department of Pharmacology & Toxicology (J.C.D., R.K.M., M.I.D., J.E.S., H.I.A., L.D.O., A.H.L.), and Department of Medicinal Chemistry (A.H.L.), Virginia Commonwealth University, Richmond, Virginia; Department of Pharmaceutical Sciences, University of Connecticut, Mansfield, Connecticut (D.A.K.); and Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas (Z.W., D.L.)
| | - Lesley D O'Brien
- Department of Pharmacology & Toxicology (J.C.D., R.K.M., M.I.D., J.E.S., H.I.A., L.D.O., A.H.L.), and Department of Medicinal Chemistry (A.H.L.), Virginia Commonwealth University, Richmond, Virginia; Department of Pharmaceutical Sciences, University of Connecticut, Mansfield, Connecticut (D.A.K.); and Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas (Z.W., D.L.)
| | - Debra A Kendall
- Department of Pharmacology & Toxicology (J.C.D., R.K.M., M.I.D., J.E.S., H.I.A., L.D.O., A.H.L.), and Department of Medicinal Chemistry (A.H.L.), Virginia Commonwealth University, Richmond, Virginia; Department of Pharmaceutical Sciences, University of Connecticut, Mansfield, Connecticut (D.A.K.); and Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas (Z.W., D.L.)
| | - Zhixing Wu
- Department of Pharmacology & Toxicology (J.C.D., R.K.M., M.I.D., J.E.S., H.I.A., L.D.O., A.H.L.), and Department of Medicinal Chemistry (A.H.L.), Virginia Commonwealth University, Richmond, Virginia; Department of Pharmaceutical Sciences, University of Connecticut, Mansfield, Connecticut (D.A.K.); and Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas (Z.W., D.L.)
| | - Dai Lu
- Department of Pharmacology & Toxicology (J.C.D., R.K.M., M.I.D., J.E.S., H.I.A., L.D.O., A.H.L.), and Department of Medicinal Chemistry (A.H.L.), Virginia Commonwealth University, Richmond, Virginia; Department of Pharmaceutical Sciences, University of Connecticut, Mansfield, Connecticut (D.A.K.); and Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas (Z.W., D.L.)
| | - Aron H Lichtman
- Department of Pharmacology & Toxicology (J.C.D., R.K.M., M.I.D., J.E.S., H.I.A., L.D.O., A.H.L.), and Department of Medicinal Chemistry (A.H.L.), Virginia Commonwealth University, Richmond, Virginia; Department of Pharmaceutical Sciences, University of Connecticut, Mansfield, Connecticut (D.A.K.); and Department of Pharmaceutical Sciences, Texas A&M, College Station, Texas (Z.W., D.L.)
| |
Collapse
|
12
|
On the Biomedical Properties of Endocannabinoid Degradation and Reuptake Inhibitors: Pre-clinical and Clinical Evidence. Neurotox Res 2021; 39:2072-2097. [PMID: 34741755 DOI: 10.1007/s12640-021-00424-z] [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: 07/19/2021] [Revised: 09/14/2021] [Accepted: 09/28/2021] [Indexed: 10/19/2022]
Abstract
The endocannabinoid system (ECS) is composed of endogenous cannabinoids; components involved in their synthesis, transport, and degradation; and an expansive variety of cannabinoid receptors. Hypofunction or deregulation of the ECS is related to pathological conditions. Consequently, endogenous enhancement of endocannabinoid levels and/or regulation of their metabolism represent promising therapeutic approaches. Several major strategies have been suggested for the modulation of the ECS: (1) blocking endocannabinoids degradation, (2) inhibition of endocannabinoid cellular uptake, and (3) pharmacological modulation of cannabinoid receptors as potential therapeutic targets. Here, we focused in this review on degradation/reuptake inhibitors over cannabinoid receptor modulators in order to provide an updated synopsis of contemporary evidence advancing mechanisms of endocannabinoids as pharmacological tools with therapeutic properties for the treatment of several disorders. For this purpose, we revisited the available literature and reported the latest advances regarding the biomedical properties of fatty acid amide hydrolase and monoacylglycerol lipase inhibitors in pre-clinical and clinical studies. We also highlighted anandamide and 2-arachidonoylglycerol reuptake inhibitors with promising results in pre-clinical studies using in vitro and animal models as an outlook for future research in clinical trials.
Collapse
|
13
|
Demir Çaltekin M, Özkut MM, Çaltekin İ, Kaymak E, Çakır M, Kara M, Yalvaç ES. The protective effect of JZL184 on ovarian ischemia reperfusion injury and ovarian reserve in rats. J Obstet Gynaecol Res 2021; 47:2692-2704. [PMID: 34008304 DOI: 10.1111/jog.14859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/03/2021] [Accepted: 05/08/2021] [Indexed: 11/28/2022]
Abstract
AIM Ovarian torsion is a gynecopathology that requires emergency surgery in women. However, ischemia reperfusion injury (IRI) occurs after treatment with detorsion. This study aimed to evaluate the effects of monoacylglycerol lipase inhibitor JZL184 on ovarian IRI and ovarian reserve. METHODS Forty-eight female Wistar albino rats were divided into six groups. Group 1: Sham, Group 2: Ischemia, Group 3: ischemia/reperfusion (IR), Group 4: IR + JZL184 4 mg/kg, Group 5: IR + JZL184 16 mg/kg, Group 6: IR + vehicle (dimethyl sulfoxide). Three hours of ischemia followed by 3 h of reperfusion. Two different doses of JZL184 (4 and 16 mg/kg) were administered intraperitoneally in Group 4 and 5, 30 min before reperfusion. Ovarian IRI and ovarian reserve were evaluated in serum and tissue by using histopathological and biochemical parameters. RESULTS Treatment with JZL184 was associated with a significant increase in ovarian 2-arachidonoylglycerol and improved serum anti-Mullerian hormone, İnhibin B, primordial follicle count, and ovarian histopathological damage score (p < 0.05). JZL184 treatment significantly decreased the level of malondialdehyde, and increased superoxide dismutase enzyme activity and glutathione (GSH) levels (p < 0.05). The increased phosphorile nuclear factor-κB (Phospho-NF-κB-p65), tumor necrosis factor alpha (TNF-α), interleukin-1beta (IL-1β), transforming growth factor beta 1 (TGF-β1), and TUNEL assay immunopositivity scores in ovarian I/R injury were decreased after treatment with JZL184 (p < 0.05). CONCLUSIONS JZL184 showed significant ameliorative effects on ovarian IRI and ovarian reserve caused by IR through acting as an antioxidant, anti-inflammatory, and antiapoptotic agent. Thus, JZL184 may be a novel therapeutic agent for ovarian IRI.
Collapse
Affiliation(s)
- Melike Demir Çaltekin
- Department of Obstetrics and Gynecology, Yozgat Bozok University Faculty of Medicine, Yozgat, Turkey
| | - Mahmud Mustafa Özkut
- Department of Histology and Embryology, Yozgat Bozok University Faculty of Medicine, Yozgat, Turkey
| | - İbrahim Çaltekin
- Department of Emergency Medicine, Yozgat Bozok University Faculty of Medicine, Yozgat, Turkey
| | - Emin Kaymak
- Department of Histology and Embryology, Yozgat Bozok University Faculty of Medicine, Yozgat, Turkey
| | - Murat Çakır
- Department of Physiology, Yozgat Bozok University Faculty of Medicine, Yozgat, Turkey
| | - Mustafa Kara
- Department of Obstetrics and Gynecology, Kırşehir Ahi Evran University Faculty of Medicine, Kırşehir, Turkey
| | - Ethem Serdar Yalvaç
- Department of Obstetrics and Gynecology, Yozgat Bozok University Faculty of Medicine, Yozgat, Turkey
| |
Collapse
|
14
|
Laksmidewi AAAP, Soejitno A. Endocannabinoid and dopaminergic system: the pas de deux underlying human motivation and behaviors. J Neural Transm (Vienna) 2021; 128:615-630. [PMID: 33712975 PMCID: PMC8105194 DOI: 10.1007/s00702-021-02326-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 03/04/2021] [Indexed: 01/11/2023]
Abstract
Endocannabinoid system (ECS) has been identified ever since cannabinoid, an active substance of Cannabis, was known to interact with endogenous cannabinoid (endocannabinoid/eCB) receptors. It later turned out that eCB was more intricate than previously thought. It has a pervasive role and exerts a multitude of cellular signaling mechanisms, regulating various physiological neurotransmission pathways in the human brain, including the dopaminergic (DA) system. eCB roles toward DA system were robust, clearly delineated, and reproducible with respect to physiological as well as pathological neurochemical and neurobehavioral manifestations of DA system, particularly those involving the nigrostriatal and mesocorticolimbic pathways. The eCB–DA system regulates the basics in the Maslow’s pyramid of hierarchy of needs required for individual survival such as food and sexual activity for reproductive purpose to those of higher needs in the pyramid, including self-actualization behaviors leading to achievement and reward (e.g., academic- and/or work-related performance and achievements). It is, thus, interesting to specifically discuss the eCB–DA system, not only on the molecular level, but also its tremendous potential to be developed as a future therapeutic strategy for various neuropsychiatric problems, including obesity, drug addiction and withdrawal, pathological hypersexuality, or low motivation behaviors.
Collapse
Affiliation(s)
- A A A Putri Laksmidewi
- Neurobehavioral and Cognitive Division, Neurology Department, Faculty of Medicine, Udayana University/Sanglah Hospital, Denpasar, Bali, Indonesia.
| | - Andreas Soejitno
- Neurobehavioral and Cognitive Division, Neurology Department, Faculty of Medicine, Udayana University/Sanglah Hospital, Denpasar, Bali, Indonesia
| |
Collapse
|
15
|
Everett TJ, Gomez DM, Hamilton LR, Oleson EB. Endocannabinoid modulation of dopamine release during reward seeking, interval timing, and avoidance. Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110031. [PMID: 32663486 DOI: 10.1016/j.pnpbp.2020.110031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/15/2020] [Accepted: 06/28/2020] [Indexed: 01/02/2023]
Abstract
Endocannabinoids (eCBs) are neuromodulators that influence a wide range of neural systems and behaviors. In the current review, we describe our recent research showing how eCBs, particularly 2-arachidonoylglycerol (2-AG), concurrently shape mesolimbic dopamine (DA) release and associated behavior. We will restrict our discussion by emphasizing three distinct behaviors: reward seeking, interval timing, and active avoidance. During reward seeking we find that 2-AG is necessary to observe cue-evoked DA release events that are thought to represent the value of a rewarding outcome. We then describe data showing that 2-AG modulates unique patterns of DA release and behavior observed under conditions of periodic reinforcement. These data are discussed within the context of interval timing and adjunctive behavior. eCB modulation of DA release is also implicated in defensive behavior, including the avoidance of harm. As in reward seeking, our data suggest that the concentration of DA that is evoked by a warning signal can represent the value of an avoidance outcome. And, disrupting eCB signaling concomitantly reduces the concentration of the avoidance value signal and active avoidance. Disruptions in reward seeking, interval timing, and defensive behavior are commonly observed in a variety of movement disorders (e.g., Parkinson's and Huntington's disease) and disorders of motivation (e.g., addiction). We believe our data on eCB-DA interactions have implications for the development of novel pharmacotherapies to treat these disorders. Thus, we conclude by discussing how eCB pharmacology might be harnessed to treat disorders of movement and motivation.
Collapse
Affiliation(s)
| | - Devan M Gomez
- Psychology Department, University of Colorado Denver, USA; Department of Biomedical Sciences, Marquette University, USA
| | | | - Erik B Oleson
- Psychology Department, University of Colorado Denver, USA; Integrative Biology Department, University of Colorado Denver, USA.
| |
Collapse
|
16
|
Bergeria CL, Huhn AS, Dunn KE. The impact of naturalistic cannabis use on self-reported opioid withdrawal. J Subst Abuse Treat 2020; 113:108005. [PMID: 32359667 PMCID: PMC7212528 DOI: 10.1016/j.jsat.2020.108005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/25/2020] [Accepted: 03/28/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Four states have legalized medical cannabis for the purpose of treating opioid use disorder. It is unclear whether cannabinoids improve or exacerbate opioid withdrawal. A more thorough examination of cannabis and its impact on specific symptoms of opioid withdrawal is warranted. METHOD Two hundred individuals recruited through Amazon Mechanical Turk with past month opioid and cannabis use and experience of opioid withdrawal completed the survey. Participants indicated which opioid withdrawal symptoms improved or worsened with cannabis use and indicated the severity of their opioid withdrawal on days with and without cannabis. RESULTS 62.5% (n = 125) of 200 participants had used cannabis to treat withdrawal. Participants most frequently indicated that cannabis improved: anxiety, tremors, and trouble sleeping. A minority of participants (6.0%, n = 12) indicated cannabis worsened opioid withdrawal, specifically symptoms of yawning, teary eyes, and runny nose. Across all symptoms, more participants indicated that symptoms improved with cannabis compared to those that indicated symptoms worsened with cannabis. Women reported greater relief from withdrawal with cannabis use than men. DISCUSSION These results show that cannabis may improve opioid withdrawal symptoms and that the size of the effect is clinically meaningful. It is important to note that symptoms are exacerbated with cannabis in only a minority of individuals. Prospectively designed studies examining the impact of cannabis and cannabinoids on opioid withdrawal are warranted.
Collapse
Affiliation(s)
- Cecilia L Bergeria
- Behavioral Pharmacology Research Unit, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, United States of America.
| | - Andrew S Huhn
- Behavioral Pharmacology Research Unit, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, United States of America
| | - Kelly E Dunn
- Behavioral Pharmacology Research Unit, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, United States of America
| |
Collapse
|
17
|
Butler K, Le Foll B. Novel therapeutic and drug development strategies for tobacco use disorder: endocannabinoid modulation. Expert Opin Drug Discov 2020; 15:1065-1080. [DOI: 10.1080/17460441.2020.1767581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Kevin Butler
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
- Acute Care Program, Centre for Addiction and Mental Health, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
Slivicki RA, Iyer V, Mali SS, Garai S, Thakur GA, Crystal JD, Hohmann AG. Positive Allosteric Modulation of CB 1 Cannabinoid Receptor Signaling Enhances Morphine Antinociception and Attenuates Morphine Tolerance Without Enhancing Morphine- Induced Dependence or Reward. Front Mol Neurosci 2020; 13:54. [PMID: 32410959 PMCID: PMC7199816 DOI: 10.3389/fnmol.2020.00054] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/17/2020] [Indexed: 01/09/2023] Open
Abstract
Opioid analgesics represent a critical treatment for chronic pain in the analgesic ladder of the World Health Organization. However, their use can result in a number of unwanted side-effects including incomplete efficacy, constipation, physical dependence, and overdose liability. Cannabinoids enhance the pain-relieving effects of opioids in preclinical studies and dampen unwanted side-effects resulting from excessive opioid intake. We recently reported that a CB1 positive allosteric modulator (PAM) exhibits antinociceptive efficacy in models of pathological pain and lacks the adverse side effects of direct CB1 receptor activation. In the present study, we evaluated whether a CB1 PAM would enhance morphine’s therapeutic efficacy in an animal model of chemotherapy-induced neuropathic pain and characterized its impact on unwanted side-effects associated with chronic opioid administration. In paclitaxel-treated mice, both the CB1 PAM GAT211 and the opioid analgesic morphine reduced paclitaxel-induced behavioral hypersensitivities to mechanical and cold stimulation in a dose-dependent manner. Isobolographic analysis revealed that combinations of GAT211 and morphine resulted in anti-allodynic synergism. In paclitaxel-treated mice, a sub-threshold dose of GAT211 prevented the development of tolerance to the anti-allodynic effects of morphine over 20 days of once daily dosing. However, GAT211 did not reliably alter somatic withdrawal signs (i.e., jumps, paw tremors) in morphine-dependent neuropathic mice challenged with naloxone. In otherwise naïve mice, GAT211 also prolonged antinociceptive efficacy of morphine in the tail-flick test and reduced the overall right-ward shift in the ED50 for morphine to produce antinociception in the tail-flick test, consistent with attenuation of morphine tolerance. Pretreatment with GAT211 did not alter somatic signs of μ opioid receptor dependence in mice rendered dependent upon morphine via subcutaneous implantation of a morphine pellet. Moreover, GAT211 did not reliably alter μ-opioid receptor-mediated reward as measured by conditioned place preference to morphine. Our results suggest that a CB1 PAM may be beneficial in enhancing and prolonging the therapeutic properties of opioids while potentially sparing unwanted side-effects (e.g., tolerance) that occur with repeated opioid treatment.
Collapse
Affiliation(s)
- Richard A Slivicki
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Vishakh Iyer
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Sonali S Mali
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Sumanta Garai
- Center for Drug Discovery, Bouve College of Health Sciences, Northeastern University, Boston, MA, United States
| | - Ganesh A Thakur
- Center for Drug Discovery, Bouve College of Health Sciences, Northeastern University, Boston, MA, United States
| | - Jonathon D Crystal
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Andrea G Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States.,Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States
| |
Collapse
|
19
|
Ross JA, Van Bockstaele EJ. The role of catecholamines in modulating responses to stress: Sex-specific patterns, implications, and therapeutic potential for post-traumatic stress disorder and opiate withdrawal. Eur J Neurosci 2020; 52:2429-2465. [PMID: 32125035 DOI: 10.1111/ejn.14714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 01/15/2020] [Accepted: 02/20/2020] [Indexed: 12/22/2022]
Abstract
Emotional arousal is one of several factors that determine the strength of a memory and how efficiently it may be retrieved. The systems at play are multifaceted; on one hand, the dopaminergic mesocorticolimbic system evaluates the rewarding or reinforcing potential of a stimulus, while on the other, the noradrenergic stress response system evaluates the risk of threat, commanding attention, and engaging emotional and physical behavioral responses. Sex-specific patterns in the anatomy and function of the arousal system suggest that sexually divergent therapeutic approaches may be advantageous for neurological disorders involving arousal, learning, and memory. From the lens of the triple network model of psychopathology, we argue that post-traumatic stress disorder and opiate substance use disorder arise from maladaptive learning responses that are perpetuated by hyperarousal of the salience network. We present evidence that catecholamine-modulated learning and stress-responsive circuitry exerts substantial influence over the salience network and its dysfunction in stress-related psychiatric disorders, and between the sexes. We discuss the therapeutic potential of targeting the endogenous cannabinoid system; a ubiquitous neuromodulator that influences learning, memory, and responsivity to stress by influencing catecholamine, excitatory, and inhibitory synaptic transmission. Relevant preclinical data in male and female rodents are integrated with clinical data in men and women in an effort to understand how ideal treatment modalities between the sexes may be different.
Collapse
Affiliation(s)
- Jennifer A Ross
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Elisabeth J Van Bockstaele
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
20
|
Dunn KE, Huhn AS, Bergeria CL, Gipson CD, Weerts EM. Non-Opioid Neurotransmitter Systems that Contribute to the Opioid Withdrawal Syndrome: A Review of Preclinical and Human Evidence. J Pharmacol Exp Ther 2019; 371:422-452. [PMID: 31391211 PMCID: PMC6863456 DOI: 10.1124/jpet.119.258004] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/30/2019] [Indexed: 01/15/2023] Open
Abstract
Opioid misuse and abuse is a major international public health issue. Opioid use disorder (OUD) is largely maintained by a desire to suppress aversive opioid withdrawal symptoms. Opioid withdrawal in patients seeking abstinence from illicit or prescribed opioids is often managed by provision of a μ-opioid agonist/partial agonist in combination with concomitant medications. Concomitant medications are administered based on their ability to treat specific symptoms rather than a mechanistic understanding of the opioid withdrawal syndrome; however, their use has not been statistically associated with improved treatment outcomes. Understanding the central and/or peripheral mechanisms that underlie individual withdrawal symptom expression in humans will help promote medication development for opioid withdrawal management. To support focused examination of mechanistically supported concomitant medications, this review summarizes evidence from preclinical (N = 68) and human (N = 30) studies that administered drugs acting on the dopamine, serotonin, cannabinoid, orexin/hypocretin, and glutamate systems and reported outcomes related to opioid withdrawal. These studies provide evidence that each of these systems contribute to opioid withdrawal severity. The Food and Drug Administration has approved medications acting on these respective systems for other indications and research in this area could support the repurposing of these medications to enhance opioid withdrawal treatment. These data support a focused examination of mechanistically informed concomitant medications to help reduce opioid withdrawal severity and enhance the continuum of care available for persons with OUD.
Collapse
Affiliation(s)
- Kelly E Dunn
- Behavioral Pharmacology Research Unit, Johns Hopkins University School of Medicine, Baltimore, Maryland (K.D.E., A.S.H., C.L.B., E.M.W.); and Arizona State University, Tempe, Arizona (C.D.G.)
| | - Andrew S Huhn
- Behavioral Pharmacology Research Unit, Johns Hopkins University School of Medicine, Baltimore, Maryland (K.D.E., A.S.H., C.L.B., E.M.W.); and Arizona State University, Tempe, Arizona (C.D.G.)
| | - Cecilia L Bergeria
- Behavioral Pharmacology Research Unit, Johns Hopkins University School of Medicine, Baltimore, Maryland (K.D.E., A.S.H., C.L.B., E.M.W.); and Arizona State University, Tempe, Arizona (C.D.G.)
| | - Cassandra D Gipson
- Behavioral Pharmacology Research Unit, Johns Hopkins University School of Medicine, Baltimore, Maryland (K.D.E., A.S.H., C.L.B., E.M.W.); and Arizona State University, Tempe, Arizona (C.D.G.)
| | - Elise M Weerts
- Behavioral Pharmacology Research Unit, Johns Hopkins University School of Medicine, Baltimore, Maryland (K.D.E., A.S.H., C.L.B., E.M.W.); and Arizona State University, Tempe, Arizona (C.D.G.)
| |
Collapse
|
21
|
Nguyen JD, Grant Y, Creehan KM, Hwang CS, Vandewater SA, Janda KD, Cole M, Taffe MA. Δ 9-tetrahydrocannabinol attenuates oxycodone self-administration under extended access conditions. Neuropharmacology 2019; 151:127-135. [PMID: 30980837 DOI: 10.1016/j.neuropharm.2019.04.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/05/2019] [Accepted: 04/08/2019] [Indexed: 11/24/2022]
Abstract
Growing nonmedical use of prescription opioids is a global problem, motivating research on ways to reduce use and combat addiction. Medical cannabis ("medical marijuana") legalization has been associated epidemiologically with reduced opioid harms and cannabinoids have been shown to modulate effects of opioids in animal models. This study was conducted to determine if Δ9-tetrahydrocannabinol (THC) enhances the behavioral effects of oxycodone. Male rats were trained to intravenously self-administer (IVSA) oxycodone (0.15 mg/kg/infusion) during 1 h, 4 h or 8 h sessions. Following acquisition rats were exposed to THC by vapor inhalation (1 h and 8 h groups) or injection (0-10 mg/kg, i.p.; all groups) prior to IVSA sessions. Fewer oxycodone infusions were obtained by rats following vaporized or injected THC compared with vehicle treatment prior to the session. Follow-up studies demonstrated parallel dose-dependent effects of THC, i.p., on self-administration of different per-infusion doses of oxycodone and a preserved loading dose early in the session. These patterns are inconsistent with behavioral suppression. Additional groups of male and female Wistar rats were assessed for nociception following inhalation of vaporized THC (50 mg/mL), oxycodone (100 mg/mL) or the combination. Tail withdrawal latency was increased more by the THC/oxycodone combination compared to either drug alone. Similar additive antinociceptive effects were produced by injection of THC (5.0 mg/kg, i.p.) and oxycodone (2.0 mg/kg, s.c.). Together these data demonstrate additive effects of THC and oxycodone and suggest the potential use of THC to enhance therapeutic efficacy, and to reduce the abuse, of opioids.
Collapse
Affiliation(s)
- Jacques D Nguyen
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Yanabel Grant
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Kevin M Creehan
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Candy S Hwang
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, CA, USA; Department of Chemistry, Southern Connecticut State University, New Haven, CT, USA
| | - Sophia A Vandewater
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Kim D Janda
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, CA, USA
| | - Maury Cole
- La Jolla Alcohol Research, Inc, La Jolla, CA, USA
| | - Michael A Taffe
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
22
|
Scherma M, Masia P, Satta V, Fratta W, Fadda P, Tanda G. Brain activity of anandamide: a rewarding bliss? Acta Pharmacol Sin 2019; 40:309-323. [PMID: 30050084 DOI: 10.1038/s41401-018-0075-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/20/2018] [Indexed: 12/11/2022] Open
Abstract
Anandamide is a lipid mediator that acts as an endogenous ligand of CB1 receptors. These receptors are also the primary molecular target responsible for the pharmacological effects of Δ9-tetrahydrocannabinol, the psychoactive ingredient in Cannabis sativa. Several studies demonstrate that anandamide exerts an overall modulatory effect on the brain reward circuitry. Several reports suggest its involvement in the addiction-producing actions of other abused drugs, and it can also act as a behavioral reinforcer in animal models of drug abuse. Importantly, all these effects of anandamide appear to be potentiated by pharmacological inhibition of its metabolic degradation. Enhanced brain levels of anandamide after treatment with inhibitors of fatty acid amide hydrolase, the main enzyme responsible for its degradation, seem to affect the rewarding and reinforcing actions of many drugs of abuse. In this review, we will provide an overview from a preclinical perspective of the current state of knowledge regarding the behavioral pharmacology of anandamide, with a particular emphasis on its motivational/reinforcing properties. We will also discuss how modulation of anandamide levels through inhibition of enzymatic metabolic pathways could provide a basis for developing new pharmaco-therapeutic tools for the treatment of substance use disorders.
Collapse
|
23
|
Di Marzo V. New approaches and challenges to targeting the endocannabinoid system. Nat Rev Drug Discov 2018; 17:623-639. [DOI: 10.1038/nrd.2018.115] [Citation(s) in RCA: 256] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Gil-Ordóñez A, Martín-Fontecha M, Ortega-Gutiérrez S, López-Rodríguez ML. Monoacylglycerol lipase (MAGL) as a promising therapeutic target. Biochem Pharmacol 2018; 157:18-32. [PMID: 30059673 DOI: 10.1016/j.bcp.2018.07.036] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/25/2018] [Indexed: 12/31/2022]
Abstract
Monoacylglycerol lipase (MAGL) has been characterized as the main enzyme responsible for the inactivation of the most abundant brain endocannabinoid, 2-arachidonoylglycerol (2-AG). Besides this role, MAGL has progressively acquired a growing importance as an integrative metabolic hub that controls not only the in vivo levels of 2-AG but also of other monoacylglycerides and, indirectly, the levels of free fatty acids derived from their hydrolysis as well as other lipids with pro-inflammatory or pro-tumorigenic effects, coming from the further metabolism of fatty acids. All these functions have only started to be elucidated in the last years due to the progress made in the knowledge of the structure of MAGL and in the development of genetic and chemical tools. In this review we report the advances made in the field with a special focus on the last decade and how MAGL has become a promising therapeutic target for the treatment of several diseases that currently lack appropriate therapies.
Collapse
Affiliation(s)
- Ana Gil-Ordóñez
- Department of Organic Chemistry, School of Chemistry, Universidad Complutense de Madrid, Av. Complutense s/n, E-28040 Madrid, Spain
| | - Mar Martín-Fontecha
- Department of Organic Chemistry, School of Chemistry, Universidad Complutense de Madrid, Av. Complutense s/n, E-28040 Madrid, Spain
| | - Silvia Ortega-Gutiérrez
- Department of Organic Chemistry, School of Chemistry, Universidad Complutense de Madrid, Av. Complutense s/n, E-28040 Madrid, Spain
| | - María L López-Rodríguez
- Department of Organic Chemistry, School of Chemistry, Universidad Complutense de Madrid, Av. Complutense s/n, E-28040 Madrid, Spain.
| |
Collapse
|
25
|
Sloan ME, Gowin JL, Ramchandani VA, Hurd YL, Le Foll B. The endocannabinoid system as a target for addiction treatment: Trials and tribulations. Neuropharmacology 2017; 124:73-83. [PMID: 28564576 DOI: 10.1016/j.neuropharm.2017.05.031] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 12/19/2022]
Abstract
Addiction remains a major public health concern, and while pharmacotherapies can be effective, clinicians are limited by the paucity of existing interventions. Endocannabinoid signaling is involved in reward and addiction, which raises the possibility that drugs targeting this system could be used to treat substance use disorders. This review discusses findings from randomized controlled trials evaluating cannabinergic medications for addiction. Current evidence suggests that pharmacotherapies containing delta-9-tetrahydrocannabinol, such as dronabinol and nabiximols, are effective for cannabis withdrawal. Dronabinol may also reduce symptoms of opioid withdrawal. The cannabinoid receptor 1 (CB1) inverse agonist rimonabant showed promising effects for smoking cessation but also caused psychiatric side effects and currently lacks regulatory approval. Few trials have investigated cannabinergic medications for alcohol use disorder. Overall, the endocannabinoid system remains a promising target for addiction treatment. Development of novel medications such as fatty acid amide hydrolase inhibitors and neutral CB1 antagonists promises to extend the range of available interventions. This article is part of the Special Issue entitled "A New Dawn in Cannabinoid Neurobiology".
Collapse
Affiliation(s)
- Matthew E Sloan
- Section on Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, 10 Center Drive (10CRC, 2-2352), Bethesda, MD, 20892-1540, USA
| | - Joshua L Gowin
- Section on Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, 10 Center Drive (10CRC, 2-2352), Bethesda, MD, 20892-1540, USA
| | - Vijay A Ramchandani
- Section on Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, 10 Center Drive (10CRC, 2-2352), Bethesda, MD, 20892-1540, USA
| | - Yasmin L Hurd
- Departments of Psychiatry, Neuroscience, Pharmacology, and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, Hess CSM Building, Floor 10, Rm 105, Office 1470, Madison Avenue, New York, NY, 10029, USA
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, Ontario, M5S 2S1, Canada; Addiction Medicine Service, Ambulatory Care and Structured Treatments, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Departments of Family and Community Medicine, Pharmacology and Toxicology, and Psychiatry, Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| |
Collapse
|
26
|
Marcus DJ, Henderson-Redmond AN, Gonek M, Zee ML, Farnsworth JC, Amin RA, Andrews MJ, Davis BJ, Mackie K, Morgan DJ. Mice expressing a "hyper-sensitive" form of the CB1 cannabinoid receptor (CB1) show modestly enhanced alcohol preference and consumption. PLoS One 2017; 12:e0174826. [PMID: 28426670 PMCID: PMC5398885 DOI: 10.1371/journal.pone.0174826] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/15/2017] [Indexed: 11/17/2022] Open
Abstract
We recently characterized S426A/S430A mutant mice expressing a desensitization-resistant form of the CB1 receptor. These mice display an enhanced response to endocannabinoids and ∆9-THC. In this study, S426A/S430A mutants were used as a novel model to test whether ethanol consumption, morphine dependence, and reward for these drugs are potentiated in mice with a "hyper-sensitive" form of CB1. Using an unlimited-access, two-bottle choice, voluntary drinking paradigm, S426A/S430A mutants exhibit modestly increased intake and preference for low (6%) but not higher concentrations of ethanol. S426A/S430A mutants and wild-type mice show similar taste preference for sucrose and quinine, exhibit normal sensitivity to the hypothermic and ataxic effects of ethanol, and have normal blood ethanol concentrations following administration of ethanol. S426A/S430A mutants develop robust conditioned place preference for ethanol (2 g/kg), morphine (10 mg/kg), and cocaine (10 mg/kg), demonstrating that drug reward is not changed in S426A/S430A mutants. Precipitated morphine withdrawal is also unchanged in opioid-dependent S426A/S430A mutant mice. Although ethanol consumption is modestly changed by enhanced CB1 signaling, reward, tolerance, and acute sensitivity to ethanol and morphine are normal in this model.
Collapse
Affiliation(s)
- David J. Marcus
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
- Department of Anesthesiology, Penn State University College of Medicine, Hershey, PA, United States of America
| | - Angela N. Henderson-Redmond
- Department of Anesthesiology, Penn State University College of Medicine, Hershey, PA, United States of America
| | - Maciej Gonek
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Michael L. Zee
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
- Department of Anesthesiology, Penn State University College of Medicine, Hershey, PA, United States of America
| | - Jill C. Farnsworth
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Randa A. Amin
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Mary-Jeanette Andrews
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Brian J. Davis
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Ken Mackie
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Daniel J. Morgan
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
- Department of Anesthesiology, Penn State University College of Medicine, Hershey, PA, United States of America
| |
Collapse
|
27
|
Grabner GF, Zimmermann R, Schicho R, Taschler U. Monoglyceride lipase as a drug target: At the crossroads of arachidonic acid metabolism and endocannabinoid signaling. Pharmacol Ther 2017; 175:35-46. [PMID: 28213089 DOI: 10.1016/j.pharmthera.2017.02.033] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Monoglyerides (MGs) are short-lived, intermediary lipids deriving from the degradation of phospho- and neutral lipids, and monoglyceride lipase (MGL), also designated as monoacylglycerol lipase (MAGL), is the major enzyme catalyzing the hydrolysis of MGs into glycerol and fatty acids. This distinct function enables MGL to regulate a number of physiological and pathophysiological processes since both MGs and fatty acids can act as signaling lipids or precursors thereof. The most prominent MG species acting as signaling lipid is 2-arachidonoyl glycerol (2-AG) which is the most abundant endogenous agonist of cannabinoid receptors in the body. Importantly, recent observations demonstrate that 2-AG represents a quantitatively important source for arachidonic acid, the precursor of prostaglandins and other inflammatory mediators. Accordingly, MGL-mediated 2-AG degradation affects lipid signaling by cannabinoid receptor-dependent and independent mechanisms. Recent genetic and pharmacological studies gave important insights into MGL's role in (patho-)physiological processes, and the enzyme is now considered as a promising drug target for a number of disorders including cancer, neurodegenerative and inflammatory diseases. This review summarizes the basics of MG (2-AG) metabolism and provides an overview on the therapeutic potential of MGL.
Collapse
Affiliation(s)
- Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria
| | - Rudolf Schicho
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria.
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| |
Collapse
|
28
|
Fox ME, Rodeberg NT, Wightman RM. Reciprocal Catecholamine Changes during Opiate Exposure and Withdrawal. Neuropsychopharmacology 2017; 42:671-681. [PMID: 27461081 PMCID: PMC5240169 DOI: 10.1038/npp.2016.135] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/28/2016] [Accepted: 07/19/2016] [Indexed: 11/09/2022]
Abstract
Dysregulated catecholamine signaling has long been implicated in drug abuse. Although much is known about adaptations following chronic drug administration, little work has investigated how a single drug exposure paired with withdrawal influences catecholamine signaling in vivo. We used fast-scan cyclic voltammetry in freely moving rats to measure real-time catecholamine overflow during acute morphine exposure and naloxone-precipitated withdrawal in two regions associated with the addiction cycle: the dopamine-dense nucleus accumbens (NAc) and norepinephrine-rich ventral bed nucleus of the stria terminalis (vBNST). We compared dopamine transients in the NAc with norepinephrine concentration changes in the vBNST, and correlated release with specific withdrawal-related behaviors. Morphine increased dopamine transients in the NAc, but did not elicit norepinephrine responses in the vBNST. Conversely, dopamine output was decreased during withdrawal, while norepinephrine was released in the vBNST during specific withdrawal symptoms. Both norepinephrine and withdrawal symptoms could be elicited in the absence of morphine by administering naloxone with an α2 antagonist. The data support reciprocal roles for dopamine and norepinephrine signaling during drug exposure and withdrawal. The data also support the allostasis model and show that negative-reinforcement may begin working after a single exposure/withdrawal episode.
Collapse
Affiliation(s)
- Megan E Fox
- Department of Chemistry and Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nathan T Rodeberg
- Department of Chemistry and Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - R Mark Wightman
- Department of Chemistry and Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA,Department of Chemistry and Neuroscience Center, University of North Carolina, Venable Hall, South Road, Chapel Hill, NC 27599, USA, Tel: +1 919 9621472, Fax: +1 919 962 2388, E-mail:
| |
Collapse
|
29
|
Wilkerson JL, Ghosh S, Mustafa M, Abdullah RA, Niphakis MJ, Cabrera R, Maldonado RL, Cravatt BF, Lichtman AH. The endocannabinoid hydrolysis inhibitor SA-57: Intrinsic antinociceptive effects, augmented morphine-induced antinociception, and attenuated heroin seeking behavior in mice. Neuropharmacology 2016; 114:156-167. [PMID: 27890602 DOI: 10.1016/j.neuropharm.2016.11.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 10/11/2016] [Accepted: 11/21/2016] [Indexed: 12/18/2022]
Abstract
Although opioids are highly efficacious analgesics, their abuse potential and other untoward side effects diminish their therapeutic utility. The addition of non-opioid analgesics offers a promising strategy to reduce required antinociceptive opioid doses that concomitantly reduce opioid-related side effects. Inhibitors of the primary endocannabinoid catabolic enzymes fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) show opioid-sparing effects in preclinical models of pain. As simultaneous inhibition of these enzymes elicits enhanced antinociceptive effects compared with single enzyme inhibition, the present study tested whether the dual FAAH-MAGL inhibitor SA-57 [4-[2-(4-chlorophenyl)ethyl]-1-piperidinecarboxylic acid 2-(methylamino)-2-oxoethyl ester] produces morphine-sparing antinociceptive effects, without major side effects associated with either drug class. SA-57 dose-dependently reversed mechanical allodynia in the constriction injury (CCI) of the sciatic nerve model of neuropathic pain and carrageenan inflammatory pain model. As previously reported, SA-57 was considerably more potent in elevating anandamide (AEA) than 2-arachidonyl glycerol (2-AG) in brain. Its anti-allodynic effects required cannabinoid (CB)1 and CB2 receptors; however, only CB2 receptors were necessary for the anti-edematous effects in the carrageenan assay. Although high doses of SA-57 alone were required to produce antinociception, low doses of this compound, which elevated AEA and did not affect 2-AG brain levels, augmented the antinociceptive effects of morphine, but lacked cannabimimetic side effects. Because of the high abuse liability of opioids and implication of the endocannabinoid system in the reinforcing effects of opioids, the final experiment tested whether SA-57 would alter heroin seeking behavior. Strikingly, SA-57 reduced heroin-reinforced nose poke behavior and the progressive ratio break point for heroin. In conclusion, the results of the present study suggest that inhibition of endocannabinoid degradative enzymes represents a promising therapeutic approach to decrease effective doses of opioids needed for clinical pain control, and may also possess therapeutic potential to reduce opioid abuse.
Collapse
Affiliation(s)
- Jenny L Wilkerson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
| | - Sudeshna Ghosh
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Mohammed Mustafa
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Rehab A Abdullah
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Micah J Niphakis
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Roberto Cabrera
- Laboratory of Neuropharmacology. Department de Ciencies Experimentals i de la Salut, Pompeu Fabra University, PRBB, C/ Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Rafael L Maldonado
- Laboratory of Neuropharmacology. Department de Ciencies Experimentals i de la Salut, Pompeu Fabra University, PRBB, C/ Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
30
|
Boggs DL, Peckham A, Boggs AA, Ranganathan M. Delta-9-tetrahydrocannabinol and cannabidiol: Separating the chemicals from the "weed," a pharmacodynamic discussion. Ment Health Clin 2016; 6:277-284. [PMID: 29955482 PMCID: PMC6007535 DOI: 10.9740/mhc.2016.11.277] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cannabis is being increasingly used as a medical treatment for a variety of illnesses. However, the cannabis plant has more than 70 different phytocannabinoids with potential pharmacologic activity. Two of the most researched phytocannabinoids are delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD). Evidence suggests CBD can decrease some of the psychomimetic effects of THC. This has led to the development of a new drug, Nabiximols, for the treatment of moderate to severe spasticity due to multiple sclerosis. A discussion of evidence supporting proposed pharmacodynamic interplay between CBD and THC is presented.
Collapse
Affiliation(s)
- Douglas Lee Boggs
- Clinical Pharmacy Specialist-Outpatient Psychiatry, VA Connecticut Healthcare System, West Haven, Connecticut; Associate Research Scientist, Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut,
| | - Alyssa Peckham
- PGY-2 Psychiatry Pharmacy Resident, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Angela A Boggs
- Clinical Pharmacy Specialist-Outpatient Psychiatry, VA Connecticut Healthcare System, Newington, Connecticut
| | - Mohini Ranganathan
- Attending Psychiatrist, VA Connecticut Healthcare System, West Haven, Connecticut; Abraham Ribicoff Research Facilities, and Assisstant Professor, Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
31
|
Ligresti A, De Petrocellis L, Di Marzo V. From Phytocannabinoids to Cannabinoid Receptors and Endocannabinoids: Pleiotropic Physiological and Pathological Roles Through Complex Pharmacology. Physiol Rev 2016; 96:1593-659. [DOI: 10.1152/physrev.00002.2016] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Apart from having been used and misused for at least four millennia for, among others, recreational and medicinal purposes, the cannabis plant and its most peculiar chemical components, the plant cannabinoids (phytocannabinoids), have the merit to have led humanity to discover one of the most intriguing and pleiotropic endogenous signaling systems, the endocannabinoid system (ECS). This review article aims to describe and critically discuss, in the most comprehensive possible manner, the multifaceted aspects of 1) the pharmacology and potential impact on mammalian physiology of all major phytocannabinoids, and not only of the most famous one Δ9-tetrahydrocannabinol, and 2) the adaptive pro-homeostatic physiological, or maladaptive pathological, roles of the ECS in mammalian cells, tissues, and organs. In doing so, we have respected the chronological order of the milestones of the millennial route from medicinal/recreational cannabis to the ECS and beyond, as it is now clear that some of the early steps in this long path, which were originally neglected, are becoming important again. The emerging picture is rather complex, but still supports the belief that more important discoveries on human physiology, and new therapies, might come in the future from new knowledge in this field.
Collapse
Affiliation(s)
- Alessia Ligresti
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Comprensorio Olivetti, Pozzuoli, Italy
| | - Luciano De Petrocellis
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Comprensorio Olivetti, Pozzuoli, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Comprensorio Olivetti, Pozzuoli, Italy
| |
Collapse
|
32
|
Lofwall MR, Babalonis S, Nuzzo PA, Elayi SC, Walsh SL. Opioid withdrawal suppression efficacy of oral dronabinol in opioid dependent humans. Drug Alcohol Depend 2016; 164:143-150. [PMID: 27234658 PMCID: PMC4910823 DOI: 10.1016/j.drugalcdep.2016.05.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 05/04/2016] [Accepted: 05/04/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND The cannabinoid (CB) system is a rational novel target for treating opioid dependence, a significant public health problem around the world. This proof-of-concept study examined the potential efficacy of a CB1 receptor partial agonist, dronabinol, in relieving signs and symptoms of opioid withdrawal. METHODS Twelve opioid dependent adults participated in this 5-week, inpatient, double-blind, randomized, placebo-controlled study. Volunteers were maintained on double-blind oxycodone (30mg oral, four times/day) and participated in a training session followed by 7 experimental sessions, each testing a single oral test dose (placebo, oxycodone 30 and 60mg, dronabinol 5, 10, 20, and 30mg [decreased from 40mg]). Placebo was substituted for oxycodone maintenance doses for 21h before each session in order to produce measurable opioid withdrawal. Outcomes included observer- and participant-ratings of opioid agonist, opioid withdrawal and psychomotor/cognitive performance. RESULTS Oxycodone produced prototypic opioid agonist effects (i.e. suppressing withdrawal and increasing subjective effects indicative of abuse liability). Dronabinol 5 and 10mg produced effects most similar to placebo, while the 20 and 30mg doses produced modest signals of withdrawal suppression that were accompanied by dose-related increases in high, sedation, bad effects, feelings of heart racing, and tachycardia. Dronabinol was not liked more than placebo, showed some impairment in cognitive performance, and was identified as marijuana with increasing dose. CONCLUSION CB1 receptor activation is a reasonable strategy to pursue for the treatment of opioid withdrawal; however, dronabinol is not a likely candidate given its modest withdrawal suppression effects of limited duration and previously reported tachycardia during opioid withdrawal.
Collapse
Affiliation(s)
- Michelle R. Lofwall
- University of Kentucky College of Medicine (UK COM), Center on Drug and Alcohol Research, 845 Angliana Ave., Lexington, KY, United States 40508,UK COM, Department of Behavioral Science, Lexington, KY, United States 40536,UK COM, Department of Psychiatry, Lexington, KY, United States 40509
| | - Shanna Babalonis
- University of Kentucky College of Medicine (UK COM), Center on Drug and Alcohol Research, 845 Angliana Ave., Lexington, KY, United States 40508,UK COM, Department of Behavioral Science, Lexington, KY, United States 40536
| | - Paul A. Nuzzo
- UK COM, Department of Behavioral Science, Lexington, KY, United States 40536
| | - Samy Claude Elayi
- UK COM, Department of Cardiology, Gill Heart Institute, Lexington, KY, United States 40536
| | - Sharon L. Walsh
- University of Kentucky College of Medicine (UK COM), Center on Drug and Alcohol Research, 845 Angliana Ave., Lexington, KY, United States 40508,UK COM, Department of Behavioral Science, Lexington, KY, United States 40536,UK COM, Department of Psychiatry, Lexington, KY, United States 40509
| |
Collapse
|
33
|
Wills KL, Parker LA. Effect of Pharmacological Modulation of the Endocannabinoid System on Opiate Withdrawal: A Review of the Preclinical Animal Literature. Front Pharmacol 2016; 7:187. [PMID: 27445822 PMCID: PMC4923145 DOI: 10.3389/fphar.2016.00187] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/13/2016] [Indexed: 01/20/2023] Open
Abstract
Over the years, animal studies have revealed a role for the endocannabinoid system in the regulation of multiple aspects of opiate addiction. The current review provides an overview of this literature in regards to opiate withdrawal. The opiate withdrawal syndrome, hypothesized to act as a negative reinforcer in mediating continued drug use, can be characterized by the emergence of spontaneous or precipitated aversive somatic and affective states following the termination of drug use. The behaviors measured to quantify somatic opiate withdrawal and the paradigms employed to assess affective opiate withdrawal (e.g., conditioned place aversion) in both acutely and chronically dependent animals are discussed in relation to the ability of the endocannabinoid system to modulate these behaviors. Additionally, the brain regions mediating somatic and affective opiate withdrawal are elucidated with respect to their modulation by the endocannabinoid system. Ultimately, a review of these findings reveals dissociations between the brain regions mediating somatic and affective opiate withdrawal, and the ability of cannabinoid type 1 (CB1) receptor agonism/antagonism to interfere with opiate withdrawal within different brain sub regions.
Collapse
Affiliation(s)
- Kiri L Wills
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph ON, Canada
| | - Linda A Parker
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph ON, Canada
| |
Collapse
|
34
|
Wills KL, Petrie GN, Millett G, Limebeer CL, Rock EM, Niphakis MJ, Cravatt BF, Parker LA. Double Dissociation of Monoacylglycerol Lipase Inhibition and CB1 Antagonism in the Central Amygdala, Basolateral Amygdala, and the Interoceptive Insular Cortex on the Affective Properties of Acute Naloxone-Precipitated Morphine Withdrawal in Rats. Neuropsychopharmacology 2016; 41:1865-73. [PMID: 26647976 PMCID: PMC4869055 DOI: 10.1038/npp.2015.356] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 11/09/2015] [Accepted: 12/02/2015] [Indexed: 02/08/2023]
Abstract
Both CB1 receptor antagonism and agonism, in particular by 2-arachidonyl glycerol (2-AG), have been shown to reduce somatic symptoms of morphine withdrawal (MWD). Here we evaluated the effects of both systemic pretreatment with the monoacylglycerol lipase (MAGL) inhibitor MJN110 (which selectively elevates 2-AG) and central administration of both MJN110 and the CB1 antagonist (AM251) on the affective properties of MWD. Acute MWD induced place aversion occurs when naloxone is administered 24 h following a single exposure to a high dose of morphine. Systemic pretreatment with the MAGL inhibitor, MJN110, prevented the aversive effects of acute MWD by a CB1 receptor-dependent mechanism. Furthermore, in a double dissociation, AM251 infusions into the central amygdala, but MJN110 infusions into the basolateral amygdala, interfered with the naloxone-precipitated MWD induced place aversion. As well, MJN110, but not AM251, infusions into the interoceptive insular cortex (a region known to be activated in acute MWD) also prevented the establishment of the place aversion by a CB1 mechanism of action. These findings reveal the respective sites of action of systemically administered MJN110 and AM251 in regulating the aversive effects of MWD.
Collapse
Affiliation(s)
- Kiri L Wills
- Collaborative Neuroscience Program, Department of Psychology, University of Guelph, Guelph, ON, Canada
| | - Gavin N Petrie
- Collaborative Neuroscience Program, Department of Psychology, University of Guelph, Guelph, ON, Canada
| | - Geneva Millett
- Collaborative Neuroscience Program, Department of Psychology, University of Guelph, Guelph, ON, Canada
| | - Cheryl L Limebeer
- Collaborative Neuroscience Program, Department of Psychology, University of Guelph, Guelph, ON, Canada
| | - Erin M Rock
- Collaborative Neuroscience Program, Department of Psychology, University of Guelph, Guelph, ON, Canada
| | - Micah J Niphakis
- Skaggs Institute for Chemical Biology, Department of Chemical Physiology, Scripps Research Institute, La Jolla, CA, USA
| | - Benjamin F Cravatt
- Skaggs Institute for Chemical Biology, Department of Chemical Physiology, Scripps Research Institute, La Jolla, CA, USA
| | - Linda A Parker
- Collaborative Neuroscience Program, Department of Psychology, University of Guelph, Guelph, ON, Canada,Collaborative Neuroscience Program, Department of Psychology, University of Guelph, 50 Stone Road E, Guelph, ON N1G 2W1, Canada, Tel: +1 519 824 4120, 5330, Fax: +1 519 837 8629, E-mail:
| |
Collapse
|
35
|
Henderson-Redmond AN, Guindon J, Morgan DJ. Roles for the endocannabinoid system in ethanol-motivated behavior. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:330-9. [PMID: 26123153 PMCID: PMC4679600 DOI: 10.1016/j.pnpbp.2015.06.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 06/15/2015] [Accepted: 06/22/2015] [Indexed: 12/19/2022]
Abstract
Alcohol use disorder represents a significant human health problem that leads to substantial loss of human life and financial cost to society. Currently available treatment options do not adequately address this human health problem, and thus, additional therapies are desperately needed. The endocannabinoid system has been shown, using animal models, to modulate ethanol-motivated behavior, and it has also been demonstrated that chronic ethanol exposure can have potentially long-lasting effects on the endocannabinoid system. For example, chronic exposure to ethanol, in either cell culture or preclinical rodent models, causes an increase in endocannabinoid levels that results in down-regulation of the cannabinoid receptor 1 (CB1) and uncoupling of this receptor from downstream G protein signaling pathways. Using positron emission tomography (PET), similar down-regulation of CB1 has been noted in multiple regions of the brain in human alcoholic patients. In rodents, treatment with the CB1 inverse agonist SR141716A (Rimonabant), or genetic deletion of CB1 leads to a reduction in voluntary ethanol drinking, ethanol-stimulated dopamine release in the nucleus accumbens, operant self-administration of ethanol, sensitization to the locomotor effects of ethanol, and reinstatement/relapse of ethanol-motivated behavior. Although the clinical utility of Rimonabant or other antagonists/inverse agonists for CB1 is limited due to negative neuropsychiatric side effects, negative allosteric modulators of CB1 and inhibitors of endocannabinoid catabolism represent therapeutic targets worthy of additional examination.
Collapse
Affiliation(s)
| | - Josée Guindon
- Department of Pharmacology and Neuroscience, Texas Tech University Health Science Center, Lubbock, TX, 79430
| | - Daniel J Morgan
- Department of Anesthesiology, Penn State University College of Medicine, Hershey, PA 17033, United States; Department of Pharmacology, Penn State University College of Medicine, Hershey, PA 17033, United States.
| |
Collapse
|
36
|
Wilkerson JL, Niphakis MJ, Grim TW, Mustafa MA, Abdullah RA, Poklis JL, Dewey WL, Akbarali H, Banks ML, Wise LE, Cravatt BF, Lichtman AH. The Selective Monoacylglycerol Lipase Inhibitor MJN110 Produces Opioid-Sparing Effects in a Mouse Neuropathic Pain Model. J Pharmacol Exp Ther 2016; 357:145-56. [PMID: 26791602 DOI: 10.1124/jpet.115.229971] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 01/19/2016] [Indexed: 12/28/2022] Open
Abstract
Serious clinical liabilities associated with the prescription of opiates for pain control include constipation, respiratory depression, pruritus, tolerance, abuse, and addiction. A recognized strategy to circumvent these side effects is to combine opioids with other antinociceptive agents. The combination of opiates with the primary active constituent of cannabis (Δ(9)-tetrahydrocannabinol) produces enhanced antinociceptive actions, suggesting that cannabinoid receptor agonists can be opioid sparing. Here, we tested whether elevating the endogenous cannabinoid 2-arachidonoylglycerol through the inhibition of its primary hydrolytic enzyme monoacylglycerol lipase (MAGL), will produce opioid-sparing effects in the mouse chronic constriction injury (CCI) of the sciatic nerve model of neuropathic pain. The dose-response relationships of i.p. administration of morphine and the selective MAGL inhibitor 2,5-dioxopyrrolidin-1-yl 4-(bis(4-chlorophenyl)methyl)piperazine-1-carboxylate (MJN110) were tested alone and in combination at equieffective doses for reversal of CCI-induced mechanical allodynia and thermal hyperalgesia. The respective ED50 doses (95% confidence interval) of morphine and MJN110 were 2.4 (1.9-3.0) mg/kg and 0.43 (0.23-0.79) mg/kg. Isobolographic analysis of these drugs in combination revealed synergistic antiallodynic effects. Acute antinociceptive effects of the combination of morphine and MJN110 required μ-opioid, CB1, and CB2 receptors. This combination did not reduce gastric motility or produce subjective cannabimimetic effects in the drug discrimination assay. Importantly, combinations of MJN110 and morphine given repeatedly (i.e., twice a day for 6 days) continued to produce antiallodynic effects with no evidence of tolerance. Taken together, these findings suggest that MAGL inhibition produces opiate-sparing events with diminished tolerance, constipation, and cannabimimetic side effects.
Collapse
Affiliation(s)
- Jenny L Wilkerson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Micah J Niphakis
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Travis W Grim
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Mohammed A Mustafa
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Rehab A Abdullah
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Justin L Poklis
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - William L Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Hamid Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Laura E Wise
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Benjamin F Cravatt
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| |
Collapse
|
37
|
Brindisi M, Brogi S, Maramai S, Grillo A, Borrelli G, Butini S, Novellino E, Allarà M, Ligresti A, Campiani G, Di Marzo V, Gemma S. Harnessing the pyrroloquinoxaline scaffold for FAAH and MAGL interaction: definition of the structural determinants for enzyme inhibition. RSC Adv 2016. [DOI: 10.1039/c6ra12524g] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The pharmacogenic pyrroloquinoxaline scaffold has been exploited for developing piperazine and 4-aminopiperidine carboxamides/carbamates as inhibitors of the endocannabinoids’ catabolic enzymes fatty acid amide hydrolase and monoacylglycerol lipase.
Collapse
|
38
|
Carey CE, Agrawal A, Zhang B, Conley ED, Degenhardt L, Heath AC, Li D, Lynskey MT, Martin NG, Montgomery GW, Wang T, Bierut LJ, Hariri AR, Nelson EC, Bogdan R. Monoacylglycerol lipase (MGLL) polymorphism rs604300 interacts with childhood adversity to predict cannabis dependence symptoms and amygdala habituation: Evidence from an endocannabinoid system-level analysis. JOURNAL OF ABNORMAL PSYCHOLOGY 2015; 124:860-77. [PMID: 26595473 PMCID: PMC4700831 DOI: 10.1037/abn0000079] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite evidence for heritable variation in cannabis involvement and the discovery of cannabinoid receptors and their endogenous ligands, no consistent patterns have emerged from candidate endocannabinoid (eCB) genetic association studies of cannabis involvement. Given interactions between eCB and stress systems and associations between childhood stress and cannabis involvement, it may be important to consider childhood adversity in the context of eCB-related genetic variation. We employed a system-level gene-based analysis of data from the Comorbidity and Trauma Study (N = 1,558) to examine whether genetic variation in six eCB genes (anabolism: DAGLA, DAGLB, NAPEPLD; catabolism: MGLL, FAAH; binding: CNR1; SNPs N = 65) and childhood sexual abuse (CSA) predict cannabis dependence symptoms. Significant interactions with CSA emerged for MGLL at the gene level (p = .009), and for rs604300 within MGLL (ΔR2 = .007, p < .001), the latter of which survived SNP-level Bonferroni correction and was significant in an additional sample with similar directional effects (N = 859; ΔR2 = .005, p = .026). Furthermore, in a third sample (N = 312), there was evidence that rs604300 genotype interacts with early life adversity to predict threat-related basolateral amygdala habituation, a neural phenotype linked to the eCB system and addiction (ΔR2 = .013, p = .047). Rs604300 may be related to epigenetic modulation of MGLL expression. These results are consistent with rodent models implicating 2-arachidonoylglycerol (2-AG), an endogenous cannabinoid metabolized by the enzyme encoded by MGLL, in the etiology of stress adaptation related to cannabis dependence, but require further replication.
Collapse
Affiliation(s)
- Caitlin E Carey
- Department of Psychology, Washington University in St. Louis
| | - Arpana Agrawal
- Department of Psychiatry, Washington University in St. Louis
| | - Bo Zhang
- Department of Genetics, Washington University in St. Louis
| | | | - Louisa Degenhardt
- National Drug and Alcohol Research Centre, University of New South Wales
| | - Andrew C Heath
- Department of Psychiatry, Washington University in St. Louis
| | - Daofeng Li
- Department of Genetics, Washington University in St. Louis
| | | | | | | | - Ting Wang
- Department of Genetics, Washington University in St. Louis
| | - Laura J Bierut
- Department of Psychiatry, Washington University in St. Louis
| | - Ahmad R Hariri
- Department of Psychology and Neuroscience, Duke University
| | - Elliot C Nelson
- Department of Psychiatry, Washington University in St. Louis
| | - Ryan Bogdan
- Department of Psychology, Washington University in St. Louis
| |
Collapse
|
39
|
Sadhasivam S, Zhang X, Chidambaran V, Mavi J, Pilipenko V, Mersha TB, Meller J, Kaufman KM, Martin LJ, McAuliffe J. Novel associations between FAAH genetic variants and postoperative central opioid-related adverse effects. THE PHARMACOGENOMICS JOURNAL 2015; 15:436-42. [PMID: 25558980 PMCID: PMC4492912 DOI: 10.1038/tpj.2014.79] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 10/15/2014] [Accepted: 11/07/2014] [Indexed: 01/01/2023]
Abstract
Opioid effects are potentiated by cannabinoid agonists including anandamide, an endocannabinoid. Inter-individual variability in responses to opioids is a major clinical problem. Multiple deaths and anoxic brain injuries occur every year because of opioid-induced respiratory depression (RD) in surgical patients and drug abusers of opioids and cannabinoids. This study aimed to determine specific associations between genetic variants of fatty acid amide hydrolase (FAAH) and postoperative central opioid adverse effects in children undergoing tonsillectomy. This is a prospective genotype-blinded observational study in which 259 healthy children between 6 and 15 years of age who received standard perioperative care with a standard anesthetic and an intraoperative dose of morphine were enrolled. Associations between frequent polymorphisms of FAAH and central postoperative opioid adverse effects including, RD, postoperative nausea and vomiting (PONV) and prolonged stay in Post Anesthesia Recovery Room (postoperative anesthesia care unit, PACU) due to RD and PONV were analyzed. Five specific FAAH single nucleotide polymorphisms (SNPs) had significant associations with more than twofold increased risk for refractory PONV (adjusted P<0.0018), and nominal associations (P<0.05) with RD and prolonged PACU stay in white children undergoing tonsillectomy. The FAAH SNP, rs324420, is a missense mutation with altered FAAH function and it is linked with other FAAH SNPs associated with PONV and RD in our cohort; association between PONV and rs324420 was confirmed in our extended cohort with additional 66 white children. Specific FAAH polymorphisms are associated with refractory PONV, opioid-related RD, and prolonged PACU stay due to opioid adverse effects in white children undergoing tonsillectomy.
Collapse
Affiliation(s)
- Senthilkumar Sadhasivam
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Xue Zhang
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Vidya Chidambaran
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Jagroop Mavi
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Valentina Pilipenko
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Tesfaye B. Mersha
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Jaroslaw Meller
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Bioinformatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kenneth M. Kaufman
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Rheumatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Cincinnati VA Medical Center, Cincinnati, OH, USA
| | - Lisa J. Martin
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - John McAuliffe
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
40
|
The potential of inhibitors of endocannabinoid metabolism as anxiolytic and antidepressive drugs--A practical view. Eur Neuropsychopharmacol 2015; 25:749-62. [PMID: 25791296 DOI: 10.1016/j.euroneuro.2015.02.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/13/2015] [Accepted: 02/20/2015] [Indexed: 12/15/2022]
Abstract
The endocannabinoid system, comprising cannabinoid CB1 and CB2 receptors, their endogenous ligands anandamide and 2-arachidonoylglyerol, and their synthetic and metabolic enzymes, are involved in many biological processes in the body, ranging from appetite to bone turnover. Compounds inhibiting the breakdown of anandamide and 2-arachidonoylglycerol increase brain levels of these lipids and thus modulate endocannabinoid signalling. In the present review, the preclinical evidence that these enzymes are good targets for development of novel therapies for anxiety and depression are discussed from a practical, rather than mechanistic, point of view. It is concluded that the preclinical data are promising, albeit tempered by problems of tolerance as well as effects upon learning and memory for irreversible monoacylglycerol lipase inhibitors, and limited by a focus upon male rodents alone. Clinical data so far has been restricted to safety studies with inhibitors of anandamide hydrolysis and a hitherto unpublished study on such a compound in elderly patients with major depressive disorders, but under the dose regimes used, they are well tolerated and show no signs of "cannabis-like" behaviours.
Collapse
|
41
|
Hruba L, Seillier A, Zaki A, Cravatt BF, Lichtman AH, Giuffrida A, McMahon LR. Simultaneous inhibition of fatty acid amide hydrolase and monoacylglycerol lipase shares discriminative stimulus effects with Δ9-tetrahydrocannabinol in mice. J Pharmacol Exp Ther 2015; 353:261-8. [PMID: 25711338 DOI: 10.1124/jpet.115.222836] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH) inhibitors exert preclinical effects indicative of therapeutic potential (i.e., analgesia). However, the extent to which MAGL and FAAH inhibitors produce unwanted effects remains unclear. Here, FAAH and MAGL inhibition was examined separately and together in a Δ(9)-tetrahydrocannabinol (Δ(9)-THC; 5.6 mg/kg i.p.) discrimination assay predictive of subjective effects associated with cannabis use, and the relative contribution of N-arachidonoyl ethanolamine (AEA) and 2-arachidonoylglycerol (2-AG) in the prefrontal cortex, hippocampus, and caudate putamen to those effects was examined. Δ(9)-THC dose-dependently increased Δ(9)-THC appropriate responses (ED50 value = 2.8 mg/kg), whereas the FAAH inhibitors PF-3845 [N-3-pyridinyl-4-[[3-[[5-(trifluoromethyl)-2-pyridinyl]oxy]phenyl]methyl]-1-piperidinecarboxamide] and URB597 [(3'-(aminocarbonyl)[1,1'-biphenyl]-3-yl)-cyclohexylcarbamate] or a MAGL inhibitor JZL184 [4-nitrophenyl-4-(dibenzo[d][1,3]dioxol-5-yl(hydroxy)methyl)piperidine-1-carboxylate] alone did not substitute for the Δ(9)-THC discriminative stimulus. The nonselective FAAH/MAGL inhibitors SA-57 [4-[2-(4-chlorophenyl)ethyl]-1-piperidinecarboxylic acid 2-(methylamino)-2-oxoethyl ester] and JZL195 [4-nitrophenyl 4-(3-phenoxybenzyl)piperazine-1-carboxylate] fully substituted for Δ(9)-THC with ED50 values equal to 2.4 and 17 mg/kg, respectively. Full substitution for Δ(9)-THC was also produced by a combination of JZL184 and PF-3845, but not by a combination of JZL184 and URB597 (i.e., 52% maximum). Cannabinoid receptor type 1 antagonist rimonabant attenuated the discriminative stimulus effects of Δ(9)-THC, SA-57, JZL195, and the combined effects of JZL184 and PF-3845. Full substitution for the Δ(9)-THC discriminative stimulus occurred only when both 2-AG and AEA were significantly elevated, and the patterns of increased endocannabinoid content were similar among brain regions. Overall, these results suggest that increasing both endogenous 2-AG and AEA produces qualitatively unique effects (i.e., the subjective effects of cannabis) that are not obtained from increasing either 2-AG or AEA separately.
Collapse
Affiliation(s)
- Lenka Hruba
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas (L.H., A.S., A.Z., A.G., L.R.M.); Department of Chemical Physiology, Skaggs Institute for Chemical Biology, Scripps Research Institute, La Jolla, California (B.F.C.); and Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (A.H.L.)
| | - Alexandre Seillier
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas (L.H., A.S., A.Z., A.G., L.R.M.); Department of Chemical Physiology, Skaggs Institute for Chemical Biology, Scripps Research Institute, La Jolla, California (B.F.C.); and Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (A.H.L.)
| | - Armia Zaki
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas (L.H., A.S., A.Z., A.G., L.R.M.); Department of Chemical Physiology, Skaggs Institute for Chemical Biology, Scripps Research Institute, La Jolla, California (B.F.C.); and Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (A.H.L.)
| | - Benjamin F Cravatt
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas (L.H., A.S., A.Z., A.G., L.R.M.); Department of Chemical Physiology, Skaggs Institute for Chemical Biology, Scripps Research Institute, La Jolla, California (B.F.C.); and Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (A.H.L.)
| | - Aron H Lichtman
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas (L.H., A.S., A.Z., A.G., L.R.M.); Department of Chemical Physiology, Skaggs Institute for Chemical Biology, Scripps Research Institute, La Jolla, California (B.F.C.); and Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (A.H.L.)
| | - Andrea Giuffrida
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas (L.H., A.S., A.Z., A.G., L.R.M.); Department of Chemical Physiology, Skaggs Institute for Chemical Biology, Scripps Research Institute, La Jolla, California (B.F.C.); and Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (A.H.L.)
| | - Lance R McMahon
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas (L.H., A.S., A.Z., A.G., L.R.M.); Department of Chemical Physiology, Skaggs Institute for Chemical Biology, Scripps Research Institute, La Jolla, California (B.F.C.); and Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (A.H.L.)
| |
Collapse
|
42
|
Befort K. Interactions of the opioid and cannabinoid systems in reward: Insights from knockout studies. Front Pharmacol 2015; 6:6. [PMID: 25698968 PMCID: PMC4318341 DOI: 10.3389/fphar.2015.00006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/08/2015] [Indexed: 12/14/2022] Open
Abstract
The opioid system consists of three receptors, mu, delta, and kappa, which are activated by endogenous opioid peptides (enkephalins, endorphins, and dynorphins). The endogenous cannabinoid system comprises lipid neuromodulators (endocannabinoids), enzymes for their synthesis and their degradation and two well-characterized receptors, cannabinoid receptors CB1 and CB2. These systems play a major role in the control of pain as well as in mood regulation, reward processing and the development of addiction. Both opioid and cannabinoid receptors are coupled to G proteins and are expressed throughout the brain reinforcement circuitry. Extending classical pharmacology, research using genetically modified mice has provided important progress in the identification of the specific contribution of each component of these endogenous systems in vivo on reward process. This review will summarize available genetic tools and our present knowledge on the consequences of gene knockout on reinforced behaviors in both systems, with a focus on their potential interactions. A better understanding of opioid-cannabinoid interactions may provide novel strategies for therapies in addicted individuals.
Collapse
Affiliation(s)
- Katia Befort
- CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives - UMR7364, Faculté de Psychologie, Neuropôle de Strasbourg - Université de Strasbourg, Strasbourg France
| |
Collapse
|
43
|
Gamage TF, Ignatowska-Jankowska BM, Muldoon PP, Cravatt BF, Damaj MI, Lichtman AH. Differential effects of endocannabinoid catabolic inhibitors on morphine withdrawal in mice. Drug Alcohol Depend 2015; 146:7-16. [PMID: 25479915 PMCID: PMC4295928 DOI: 10.1016/j.drugalcdep.2014.11.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/16/2014] [Accepted: 11/16/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Inhibition of endocannabinoid catabolic enzymes fatty acid amide hydrolase (FAAH) and/or monoacylglycerol lipase (MAGL) reduces somatic morphine withdrawal signs, but its effects on aversive aspects of withdrawal are unknown. The present study investigated whether Δ(9)-tetrahydrocannabinol (THC), the MAGL inhibitor JZL184, the FAAH inhibitor PF-3845, or the dual FAAH/MAGL inhibitor SA-57 would reduce acquisition of morphine withdrawal-induced conditioned place avoidance (CPA) and jumping. METHODS Mice were implanted with placebo or 75 mg morphine pellets, 48 h later injected with naloxone or saline and placed in the conditioning apparatus, and assessed for CPA at 72 h. Subjects were also observed for jumping behavior following naloxone challenge. RESULTS Naloxone (0.056 mg/kg) produced robust CPA in morphine-pelleted, but not placebo-pelleted, mice. Morphine pretreatment prevented the occurrence of withdrawal CPA and withdrawal jumping, while clonidine (an α2 adrenergic receptor agonist) only blocked withdrawal CPA. THC, JZL184, and SA-57 significantly reduced the percentage of mice that jumped during the conditioning session, but did not affect acquisition of withdrawal CPA. PF-3845 did not reduce morphine withdrawal CPA or jumping. Finally, neither THC nor the endocannabinoid catabolic enzyme inhibitors in non-dependent mice elicited a conditioned place preference or aversion. CONCLUSIONS These findings suggest that inhibiting endocannabinoid catabolic enzymes reduces somatic morphine withdrawal signs, but not aversive aspects as inferred in the CPA paradigm. The observation that non-dependent mice administered inhibitors of endocannabinoid degradation did not display place preferences is consistent with the idea that that endocannabinoid catabolic enzymes might be targeted therapeutically, with reduced risk of abuse.
Collapse
Affiliation(s)
- Thomas F. Gamage
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Kontos Medical Sciences Building, 1217 East Marshall Street, Richmond, VA, 23298
| | - Bogna M. Ignatowska-Jankowska
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Kontos Medical Sciences Building, 1217 East Marshall Street, Richmond, VA, 23298
| | - Pretal P. Muldoon
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Kontos Medical Sciences Building, 1217 East Marshall Street, Richmond, VA, 23298
| | - Benjamin F. Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Rd. La Jolla, CA 92037
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Kontos Medical Sciences Building, 1217 East Marshall Street, Richmond, VA, 23298
| | - Aron H. Lichtman
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Kontos Medical Sciences Building, 1217 East Marshall Street, Richmond, VA, 23298
| |
Collapse
|
44
|
The Potential of Inhibitors of Endocannabinoid Metabolism for Drug Development: A Critical Review. Handb Exp Pharmacol 2015; 231:95-128. [PMID: 26408159 DOI: 10.1007/978-3-319-20825-1_4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The endocannabinoids anandamide and 2-arachidonoylglycerol are metabolised by both hydrolytic enzymes (primarily fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MGL)) and oxygenating enzymes (e.g. cyclooxygenase-2, COX-2). In the present article, the in vivo data for compounds inhibiting endocannabinoid metabolism have been reviewed, focussing on inflammation and pain. Potential reasons for the failure of an FAAH inhibitor in a clinical trial in patients with osteoarthritic pain are discussed. It is concluded that there is a continued potential for compounds inhibiting endocannabinoid metabolism in terms of drug development, but that it is wise not to be unrealistic in terms of expectations of success.
Collapse
|
45
|
Abstract
OBJECTIVE Substance dependence disorder is a chronically relapsing condition characterised by neurobiological changes leading to loss of control in restricting a substance intake, compulsion and withdrawal syndrome. In the past few years, (endo)cannabinoids have been raised as a possible target in the aetiology of drug addiction. On the other hand, although the exact mechanisms of the genesis of addiction remain poorly understood, it is possible that neuroinflammation might also play a role in the pathophysiology of this condition. Studies demonstrated that (endo)cannabinoids act as immunomodulators by inhibiting cytokines production and microglial cell activation. Thus, in the present review, we explore the possible role of neuroinflammation on the therapeutic effects of cannabinoids on drug addiction. METHODS We conducted an evidence-based review of the literature in order to assess the role of cannabinoids on the neuroinflammatory hypothesis of addiction (terms: addiction, cannabinoids and inflammation). We searched PubMed and BioMedCentral databases up to April 2014 with no date restrictions. RESULTS In all, 165 eligible articles were included in the present review. Existing evidence suggests that disruption in cannabinoid signalling during the drug addiction process leads to microglial activation and neuroinflammation. CONCLUSION The literature showed that inflammation and changes in endocannabinod signalling occur in drug abuse; however, it remains uncertain whether these changes are causally or coincidentally associated with addiction. Additional studies, therefore, are needed to elucidate the contribution of neuroinflammation on the behavioural and neuroprotective effects of cannabinoids on drug addiction.
Collapse
|
46
|
Abstract
This paper is the thirty-sixth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2013 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia; stress and social status; tolerance and dependence; learning and memory; eating and drinking; alcohol and drugs of abuse; sexual activity and hormones, pregnancy, development and endocrinology; mental illness and mood; seizures and neurologic disorders; electrical-related activity and neurophysiology; general activity and locomotion; gastrointestinal, renal and hepatic functions; cardiovascular responses; respiration and thermoregulation; and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
47
|
Wilson-Poe AR, Lau BK, Vaughan CW. Repeated morphine treatment alters cannabinoid modulation of GABAergic synaptic transmission within the rat periaqueductal grey. Br J Pharmacol 2014; 172:681-90. [PMID: 24916363 DOI: 10.1111/bph.12809] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 05/29/2014] [Accepted: 06/05/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Cannabinoids and opioids produce antinociception by modulating GABAergic synaptic transmission in a descending analgesic pathway from the midbrain periaqueductal grey (PAG). While chronic opioid treatment produces opioid tolerance, it has recently been shown to enhance cannabinoid-induced antinociception within the PAG. This study examined the effect of repeated opioid treatment on opioid and cannabinoid presynaptic modulation of GABAergic synaptic transmission in PAG. EXPERIMENTAL APPROACH Midbrain PAG slices were prepared from untreated rats, and rats that had undergone repeated morphine or saline pretreatment. Whole-cell voltage-clamp recordings were made from neurons within the ventrolateral PAG. KEY RESULTS In slices from untreated animals, the cannabinoid receptor agonist WIN55212 and the μ receptor agonist DAMGO inhibited electrically evoked GABAA receptor-mediated inhibitory postsynaptic currents (IPSCs) IPSCs in PAG neurons, with IC50 s of 30 and 100 nM respectively. The inhibition of evoked IPSCs produced by WIN55212 (30 nM) and DAMGO (100 nM) was similar in PAG neurons from morphine- and saline-treated animals. The cannabinoid CB1 receptor antagonist AM251 increased the frequency of spontaneous miniature IPSCs in PAG neurons from repeated morphine-, but not saline-treated animals. DAMGO inhibition of evoked IPSCs was enhanced in the presence of AM251 in morphine-, but not saline-treated animals. CONCLUSIONS AND IMPLICATIONS These results indicate that the efficiency of agonist-induced inhibition of GABAergic synaptic transmission is enhanced by morphine treatment, although this is dampened by endocannabinoid-mediated tonic inhibition. Thus, endocannabinoid modulation of synaptic transmission could provide an alternative analgesic approach in a morphine-tolerant state. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- A R Wilson-Poe
- Pain Management Research Institute, Kolling Institute for Medical Research, Northern Clinical School, The University of Sydney, St Leonards, NSW, Australia
| | | | | |
Collapse
|
48
|
Oleson EB, Cachope R, Fitoussi A, Cheer JF. Tales from the dark side: do neuromodulators of drug withdrawal require changes in endocannabinoid tone? Prog Neuropsychopharmacol Biol Psychiatry 2014; 52:17-23. [PMID: 23911441 PMCID: PMC3874071 DOI: 10.1016/j.pnpbp.2013.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 07/15/2013] [Accepted: 07/23/2013] [Indexed: 10/26/2022]
Abstract
Environmental and interoceptive cues are theorized to serve as 'signals' that motivate drug seeking and effects that may be augmented in the withdrawn state. Phasic dopamine release events are observed in the nucleus accumbens in response to such motivational salient stimuli and are thought to be necessary for drug-associated cues to trigger craving. We recently demonstrated how dopamine neurons encode stimuli conditioned to a negative event, as might occur during conditioned withdrawal, and stimuli predicting the avoidance of negative events, as might occur as an addict seeks out drugs to prevent withdrawal. In this review we first discuss how the subsecond dopamine release events might process conditioned withdrawal and drug seeking driven by negative reinforcement processes within the context of our dopamine data obtained during conditioned avoidance procedures. We next describe how the endocannabinoid system modulates phasic dopamine release events and how it might be harnessed to treat negative affective states in addiction. Specifically, we have demonstrated that endocannabinoids in the ventral tegmentum sculpt cue-induced accumbal surges in dopamine release and, therefore, may also be mobilized during drug withdrawal.
Collapse
Affiliation(s)
- Erik B. Oleson
- University of Maryland, School of Medicine, 20 Penn St. Baltimore MD 21201,University of Colorado Denver, P.O. Box 173364, Denver, CO 80217
| | - Roger Cachope
- University of Maryland, School of Medicine, 20 Penn St. Baltimore MD 21201
| | - Aurelie Fitoussi
- University of Maryland, School of Medicine, 20 Penn St. Baltimore MD 21201
| | - Joseph F. Cheer
- University of Maryland, School of Medicine, 20 Penn St. Baltimore MD 21201
| |
Collapse
|
49
|
Schlosburg JE, Kinsey SG, Ignatowska-Jankowska B, Ramesh D, Abdullah RA, Tao Q, Booker L, Long JZ, Selley DE, Cravatt BF, Lichtman AH. Prolonged monoacylglycerol lipase blockade causes equivalent cannabinoid receptor type 1 receptor-mediated adaptations in fatty acid amide hydrolase wild-type and knockout mice. J Pharmacol Exp Ther 2014; 350:196-204. [PMID: 24849924 DOI: 10.1124/jpet.114.212753] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Complementary genetic and pharmacological approaches to inhibit monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH), the primary hydrolytic enzymes of the respective endogenous cannabinoids 2-arachidonoylglycerol (2-AG) and N-arachidonoylethanolamine, enable the exploration of potential therapeutic applications and physiologic roles of these enzymes. Complete and simultaneous inhibition of both FAAH and MAGL produces greatly enhanced cannabimimetic responses, including increased antinociception, and other cannabimimetic effects, far beyond those seen with inhibition of either enzyme alone. While cannabinoid receptor type 1 (CB1) function is maintained following chronic FAAH inactivation, prolonged excessive elevation of brain 2-AG levels, via MAGL inhibition, elicits both behavioral and molecular signs of cannabinoid tolerance and dependence. Here, we evaluated the consequences of a high dose of the MAGL inhibitor JZL184 [4-nitrophenyl 4-(dibenzo[d][1,3]dioxol-5-yl(hydroxy)methyl)piperidine-1-carboxylate; 40 mg/kg] given acutely or for 6 days in FAAH(-/-) and (+/+) mice. While acute administration of JZL184 to FAAH(-/-) mice enhanced the magnitude of a subset of cannabimimetic responses, repeated JZL184 treatment led to tolerance to its antinociceptive effects, cross-tolerance to the pharmacological effects of Δ(9)-tetrahydrocannabinol, decreases in CB1 receptor agonist-stimulated guanosine 5'-O-(3-[(35)S]thio)triphosphate binding, and dependence as indicated by rimonabant-precipitated withdrawal behaviors, regardless of genotype. Together, these data suggest that simultaneous elevation of both endocannabinoids elicits enhanced cannabimimetic activity but MAGL inhibition drives CB1 receptor functional tolerance and cannabinoid dependence.
Collapse
Affiliation(s)
- Joel E Schlosburg
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Steven G Kinsey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Bogna Ignatowska-Jankowska
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Divya Ramesh
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Rehab A Abdullah
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Qing Tao
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Lamont Booker
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Jonathan Z Long
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Dana E Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Benjamin F Cravatt
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| |
Collapse
|
50
|
Makriyannis A. 2012 Division of medicinal chemistry award address. Trekking the cannabinoid road: a personal perspective. J Med Chem 2014; 57:3891-911. [PMID: 24707904 DOI: 10.1021/jm500220s] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
My involvement with the field of cannabinoids spans close to 3 decades and covers a major part of my scientific career. It also reflects the robust progress in this initially largely unexplored area of biology. During this period of time, I have witnessed the growth of modern cannabinoid biology, starting from the discovery of its two receptors and followed by the characterization of its endogenous ligands and the identification of the enzyme systems involved in their biosynthesis and biotransformation. I was fortunate enough to start at the beginning of this new era and participate in a number of the new discoveries. It has been a very exciting journey. With coverage of some key aspects of my work during this period of "modern cannabinoid research," this Award Address, in part historical, intends to give an account of how the field grew, the key discoveries, and the most promising directions for the future.
Collapse
Affiliation(s)
- Alexandros Makriyannis
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University , 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|