1
|
Pakkhesal S, Shakouri M, Mosaddeghi-Heris R, Kiani Nasab S, Salehi N, Sharafi A, Ahmadalipour A. Bridging the gap: The endocannabinoid system as a functional fulcrum for benzodiazepines in a novel frontier of anxiety pharmacotherapy. Pharmacol Ther 2025; 267:108799. [PMID: 39862927 DOI: 10.1016/j.pharmthera.2025.108799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/27/2024] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
While benzodiazepines have been a mainstay of the pharmacotherapy of anxiety disorders, their short-term efficacy and risk of abuse have driven the exploration of alternative treatment approaches. The endocannabinoid (eCB) system has emerged as a key modulator of anxiety-related processes, with evidence suggesting dynamic interactions between the eCB system and the GABAergic system, the primary target of benzodiazepines. According to the existing literature, the activation of the cannabinoid receptors has been shown to exert anxiolytic effects, while their blockade or genetic deletion results in heightened anxiety-like responses. Moreover, studies have provided evidence of interactions between the eCB system and benzodiazepines in anxiety modulation. For instance, the attenuation of benzodiazepine-induced anxiolysis by cannabinoid receptor antagonism or genetic variations in the eCB system components in animal studies, have been associated with variations in benzodiazepine response and susceptibility to anxiety disorders. The combined use of cannabinoid-based medications, such as cannabinoid receptor agonists and benzodiazepine co-administration, has shown promise in augmenting anxiolytic effects and reducing benzodiazepine dosage requirements. This article aims to comprehensively review and discuss the current evidence on the involvement of the eCB system as a key modulator of benzodiazepine-related anxiolytic effects, and further, the possible mechanisms by which the region-specific eCB system-GABAergic connectivity modulates the neuro-endocrine/behavioral stress response, providing an inclusive understanding of the complex interplay between the eCB system and benzodiazepines in the context of anxiety regulation, to inform future research and clinical practice.
Collapse
Affiliation(s)
- Sina Pakkhesal
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Shakouri
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mosaddeghi-Heris
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Kiani Nasab
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Salehi
- Student Research Committee, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - AmirMohammad Sharafi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Ahmadalipour
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biomedical Engineering, The City College of New York, New York, NY, USA.
| |
Collapse
|
2
|
Ten Barge JA, van den Bosch GE, Slater R, van den Hoogen NJ, Reiss IKM, Simons SHP. Visceral Pain in Preterm Infants with Necrotizing Enterocolitis: Underlying Mechanisms and Implications for Treatment. Paediatr Drugs 2025:10.1007/s40272-024-00676-0. [PMID: 39752054 DOI: 10.1007/s40272-024-00676-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 01/04/2025]
Abstract
Necrotizing enterocolitis (NEC) is a relatively rare but very severe gastrointestinal disease primarily affecting very preterm infants. NEC is characterized by excessive inflammation and ischemia in the intestines, and is associated with prolonged, severe visceral pain. Despite its recognition as a highly painful disease, current pain management for NEC is often inadequate, and research on optimal analgesic therapy for these patients is lacking. Insight into the mechanisms underlying intestinal pain in infants with NEC-visceral pain-could help identify the most effective analgesics for these vulnerable patients. Therefore, this comprehensive review aims to provide an overview of visceral nociception, including transduction, transmission, modulation, and experience, and discuss the implications for analgesic therapy in preterm infants with NEC. The transmission of visceral pain differs from that of somatic pain, contributing to the diffuse nature of visceral pain. Studies evaluating the effectiveness of analgesics for treating visceral pain in infants are scarce. However, research in visceral pain models highlights agents that may be particularly effective for treating visceral pain based on their mechanisms of action. Further research is necessary to determine whether agents that have shown promise for treating visceral pain in preclinical studies and adults are effective in infants with NEC as well.
Collapse
Affiliation(s)
- Judith A Ten Barge
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands.
| | - Gerbrich E van den Bosch
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | - Irwin K M Reiss
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Sinno H P Simons
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Gazzi T, Brennecke B, Olikauskas V, Hochstrasser R, Wang H, Keen Chao S, Atz K, Mostinski Y, Topp A, Heer D, Kaufmann I, Ritter M, Gobbi L, Hornsperger B, Wagner B, Richter H, O'Hara F, Wittwer MB, Jul Hansen D, Collin L, Kuhn B, Benz J, Grether U, Nazaré M. Development of a Highly Selective NanoBRET Probe to Assess MAGL Inhibition in Live Cells. Chembiochem 2024:e202400704. [PMID: 39607084 DOI: 10.1002/cbic.202400704] [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: 08/27/2024] [Revised: 10/23/2024] [Indexed: 11/29/2024]
Abstract
Cell-free enzymatic assays are highly useful tools in early compound profiling due to their robustness and scalability. However, their inadequacy to reflect the complexity of target engagement in a cellular environment may lead to a significantly divergent pharmacology that is eventually observed in cells. The discrepancy that emerges from properties like permeability and unspecific protein binding may largely mislead lead compound selection to undergo further chemical optimization. We report the development of a new intracellular NanoBRET assay to assess MAGL inhibition in live cells. Based on a reverse design approach, a highly potent, reversible preclinical inhibitor was conjugated to the cell-permeable BODIPY590 acceptor fluorophore while retaining its overall balanced properties. An engineered MAGL-nanoluciferase (Nluc) fusion protein provided a suitable donor counterpart for the facile interrogation of intracellular ligand activity. Validation of assay conditions using a selection of known MAGL inhibitors set the stage for the evaluation of over 1'900 MAGL drug candidates derived from our discovery program. This evaluation enabled us to select compounds for further development based not only on target engagement, but also on favorable physicochemical parameters like permeability and protein binding. This study highlights the advantages of cell-based target engagement assays for accelerating compound profiling and progress at the early stages of drug discovery programs.
Collapse
Affiliation(s)
- Thais Gazzi
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Benjamin Brennecke
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Valentas Olikauskas
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Remo Hochstrasser
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Haiyan Wang
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Suzan Keen Chao
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Kenneth Atz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Yelena Mostinski
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Andreas Topp
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Dominik Heer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Isabelle Kaufmann
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Martin Ritter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Luca Gobbi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Benoit Hornsperger
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Bjoern Wagner
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Hans Richter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Fionn O'Hara
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Matthias B Wittwer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Dennis Jul Hansen
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Ludovic Collin
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Bernd Kuhn
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Joerg Benz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Uwe Grether
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Marc Nazaré
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| |
Collapse
|
4
|
Kuhn B, Ritter M, Hornsperger B, Bell C, Kocer B, Rombach D, Lutz MDR, Gobbi L, Kuratli M, Bartelmus C, Bürkler M, Koller R, Tosatti P, Ruf I, Guerard M, Pavlovic A, Stephanus J, O'Hara F, Wetzl D, Saal W, Stihle M, Roth D, Hug M, Huber S, Heer D, Kroll C, Topp A, Schneider M, Gertsch J, Glasmacher S, van der Stelt M, Martella A, Wittwer MB, Collin L, Benz J, Richter H, Grether U. Structure-Guided Discovery of cis-Hexahydro-pyrido-oxazinones as Reversible, Drug-like Monoacylglycerol Lipase Inhibitors. J Med Chem 2024; 67:18448-18464. [PMID: 39360636 DOI: 10.1021/acs.jmedchem.4c01769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Monoacylglycerol lipase (MAGL) is a key enzyme involved in the metabolism of the endogenous signaling ligand 2-arachidonoylglycerol, a neuroprotective endocannabinoid intimately linked to central nervous system (CNS) disorders associated with neuroinflammation. In the quest for novel MAGL inhibitors, a focused screening approach on a Roche library subset provided a reversible benzoxazinone hit exhibiting high ligand efficiency. The subsequent design of the three-dimensional cis-hexahydro-pyrido-oxazinone (cis-HHPO) moiety as benzoxazinone replacement enabled the combination of high MAGL potency with favorable ADME properties. Through enzymatic resolution an efficient synthetic route of the privileged cis-(4R,8S) HHPO headgroup was established, providing access to the highly potent and selective MAGL inhibitor 7o. Candidate molecule 7o matches the target compound profile of CNS drugs as it achieves high CSF exposures after systemic administration in rodents. It engages with the target in the brain and modulates neuroinflammatory processes, thus holding great promise for the treatment of CNS disorders.
Collapse
Affiliation(s)
- Bernd Kuhn
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Martin Ritter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Benoit Hornsperger
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Charles Bell
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Buelent Kocer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Didier Rombach
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Marius D R Lutz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Luca Gobbi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Martin Kuratli
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Christian Bartelmus
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Markus Bürkler
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Raffael Koller
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Paolo Tosatti
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Iris Ruf
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Melanie Guerard
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Anto Pavlovic
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Juliane Stephanus
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Fionn O'Hara
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Dennis Wetzl
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Wiebke Saal
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Martine Stihle
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Doris Roth
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Melanie Hug
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Sylwia Huber
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Dominik Heer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Carsten Kroll
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Andreas Topp
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Manfred Schneider
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern 3012, Switzerland
| | - Sandra Glasmacher
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern 3012, Switzerland
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University and Oncode Institute, Leiden 2300 CC, Netherlands
| | - Andrea Martella
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University and Oncode Institute, Leiden 2300 CC, Netherlands
| | - Matthias Beat Wittwer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Ludovic Collin
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Jörg Benz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Hans Richter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Uwe Grether
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| |
Collapse
|
5
|
Mallick K, Khan MF, Banerjee S. The anxiolytic effects of cannabinoids: A comprehensive review. Pharmacol Biochem Behav 2024; 243:173828. [PMID: 39032530 DOI: 10.1016/j.pbb.2024.173828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Cannabinoids, notably cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC), have emerged as promising candidates for anxiety disorder treatment, supported by both preclinical and clinical evidence. CBD exhibits notable anxiolytic effects with a favourable safety profile, though concerns regarding mild side effects and drug interactions remain. Conversely, THC, the primary psychoactive compound, presents a range of side effects, underscoring the importance of careful dosage management and individualized treatment strategies. So far there are no FDA approved cannabinoid medications for anxiety. The review highlights challenges in cannabinoid research, including dosage variability, variable preclinical data, and limited long-term data. Despite these limitations, cannabinoids represent a promising avenue for anxiety management, with the potential for further optimization in formulation, dosing protocols, and consideration of interactions with conventional therapies. Addressing these challenges could pave the way for novel and personalized approaches to treating anxiety disorders using cannabinoid-based therapies.
Collapse
Affiliation(s)
- Keya Mallick
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Mohd Faiz Khan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Sugato Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India.
| |
Collapse
|
6
|
Quintero JM, Diaz LE, Galve-Roperh I, Bustos RH, Leon MX, Beltran S, Dodd S. The endocannabinoid system as a therapeutic target in neuropathic pain: a review. Expert Opin Ther Targets 2024; 28:739-755. [PMID: 39317147 DOI: 10.1080/14728222.2024.2407824] [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: 03/06/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION This review highlights the critical role of the endocannabinoid system (ECS) in regulating neuropathic pain and explores the therapeutic potential of cannabinoids. Understanding the mechanisms of the ECS, including its receptors, endogenous ligands, and enzymatic routes, can lead to innovative treatments for chronic pain, offering more effective therapies for neuropathic conditions. This review bridges the gap between preclinical studies and clinical applications by emphasizing ECS modulation for better pain management outcomes. AREAS COVERED A review mapped the existing literature on neuropathic pain and the effects of modulating the ECS using natural and synthetic cannabinoids. This analysis examined ECS components and their alterations in neuropathic pain, highlighting the peripheral, spinal, and supraspinal mechanisms. This review aimed to provide a thorough understanding of the therapeutic potential of cannabinoids in the management of neuropathic pain. EXPERT OPINION Advances in cannabinoid research have shown significant potential for the management of chronic neuropathic pain. The study emphasizes the need for high-quality clinical trials and collaborative efforts among researchers, clinicians, and regulatory bodies to ensure safe and effective integration of cannabinoids into pain management protocols. Understanding the mechanisms and optimizing cannabinoid formulations and delivery methods are crucial for enhancing therapeutic outcomes.
Collapse
Affiliation(s)
- Jose-Manuel Quintero
- Department of Clinical Pharmacology, Evidence-Based Therapeutics Group, Faculty of Medicine, Universidad de La Sabana and Clínica Universidad de La Sabana, Chía, Cundinamarca, Colombia
- Doctoral Programme of Biosciences, Universidad de La Sabana, Chía, Colombia
| | | | - Ismael Galve-Roperh
- Department of Biochemistry and Molecular Biology, School of Chemistry and Instituto de Investigación en Neuroquímica, Complutense University, Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Rosa-Helena Bustos
- Department of Clinical Pharmacology, Evidence-Based Therapeutics Group, Faculty of Medicine, Universidad de La Sabana and Clínica Universidad de La Sabana, Chía, Cundinamarca, Colombia
| | - Marta-Ximena Leon
- Grupo Dolor y Cuidados Paliativos, Universidad de La Sabana, Chía, Colombia
| | | | - Seetal Dodd
- Faculty of Medicine, Universidad de La Sabana, Chía, Colombia
- IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Barwon Health, Geelong, Australia
- Centre for Youth Mental Health, University of Melbourne, Parkville, Australia
| |
Collapse
|
7
|
Maehashi S, Arora K, Fisher AL, Schweitzer DR, Akefe IO. Neurolipidomic insights into anxiety disorders: Uncovering lipid dynamics for potential therapeutic advances. Neurosci Biobehav Rev 2024; 163:105741. [PMID: 38838875 DOI: 10.1016/j.neubiorev.2024.105741] [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: 02/01/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
Anxiety disorders constitute a spectrum of psychological conditions affecting millions of individuals worldwide, imposing a significant health burden. Historically, the development of anxiolytic medications has been largely focused on neurotransmitter function and modulation. However, in recent years, neurolipids emerged as a prime target for understanding psychiatric pathogenesis and developing novel medications. Neurolipids influence various neural activities such as neurotransmission and cellular functioning, as well as maintaining cell membrane integrity. Therefore, this review aims to elucidate the alterations in neurolipids associated with an anxious mental state and explore their potential as targets of novel anxiolytic medications. Existing evidence tentatively associates dysregulated neurolipid levels with the etiopathology of anxiety disorders. Notably, preclinical investigations suggest that several neurolipids, including endocannabinoids and polyunsaturated fatty acids, may hold promise as potential pharmacological targets. Overall, the current literature tentatively suggests the involvement of lipids in the pathogenesis of anxiety disorders, hinting at potential prospects for future pharmacological interventions.
Collapse
Affiliation(s)
- Saki Maehashi
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
| | - Kabir Arora
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andre Lara Fisher
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | | | - Isaac Oluwatobi Akefe
- Academy for Medical Education, The University of Queensland, Herston, QLD 4006, Australia.
| |
Collapse
|
8
|
Zhao H, Liu Y, Cai N, Liao X, Tang L, Wang Y. Endocannabinoid Hydrolase Inhibitors: Potential Novel Anxiolytic Drugs. Drug Des Devel Ther 2024; 18:2143-2167. [PMID: 38882045 PMCID: PMC11179644 DOI: 10.2147/dddt.s462785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024] Open
Abstract
Over the past decade, the idea of targeting the endocannabinoid system to treat anxiety disorders has received increasing attention. Previous studies focused more on developing cannabinoid receptor agonists or supplementing exogenous cannabinoids, which are prone to various adverse effects due to their strong pharmacological activity and poor receptor selectivity, limiting their application in clinical research. Endocannabinoid hydrolase inhibitors are considered to be the most promising development strategies for the treatment of anxiety disorders. More recent efforts have emphasized that inhibition of two major endogenous cannabinoid hydrolases, monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH), indirectly activates cannabinoid receptors by increasing endogenous cannabinoid levels in the synaptic gap, circumventing receptor desensitization resulting from direct enhancement of endogenous cannabinoid signaling. In this review, we comprehensively summarize the anxiolytic effects of MAGL and FAAH inhibitors and their potential pharmacological mechanisms, highlight reported novel inhibitors or natural products, and provide an outlook on future directions in this field.
Collapse
Affiliation(s)
- Hongqing Zhao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| | - Yang Liu
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| | - Na Cai
- Outpatient Department, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Xiaolin Liao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| | - Lin Tang
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
- Department of Pharmacy, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Yuhong Wang
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
9
|
Hao Q, Shi J, Zhang Z, Yang G, Zhi Y, Wang K, Ma D, Fu S, Dong H, Zhi Z, Zhang W, Li T, Wang J. Discovery of a novel class of reversible monoacylglycerol lipase inhibitors for potential treatment of depression. Eur J Med Chem 2024; 268:116285. [PMID: 38428273 DOI: 10.1016/j.ejmech.2024.116285] [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: 01/10/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 03/03/2024]
Abstract
Biological studies on the endocannabinoid system (ECS) have suggested that monoacylglycerol lipase (MAGL), an essential enzyme responsible for the hydrolysis of 2-arachidonoylglycerol (2-AG), is a novel target for developing antidepressants. A decrease of 2-AG levels in the hippocampus of the brain has been observed in depressive-like models induced by chronic stress. Herein, employing a structure-based approach, we designed and synthesized a new class of (piperazine-1-carbonyl) quinolin-2(1H)-one derivatives as potent, reversible and selective MAGL inhibitors. And detailed structure-activity relationships (SAR) studies were discussed. Compound 27 (IC50 = 10.3 nM) exhibited high bioavailability (92.7%) and 2-AG elevation effect in vivo. Additionally, compound 27 exerted rapid antidepressant effects caused by chronic restraint stress (CRS) and didn't show signs of addictive properties in the conditioned place preference (CPP) assays. Our study is the first to report that reversible MAGL inhibitors can treat chronic stress-induced depression effectively, which may provide a new potential therapeutic strategy for the discovery of an original class of safe, rapid antidepressant drugs.
Collapse
Affiliation(s)
- Qingjing Hao
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Junwei Shi
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhilan Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Guoqing Yang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yunbao Zhi
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ke Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Dingchen Ma
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Shengnan Fu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Haijuan Dong
- The Public Laboratory Platform, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhuoer Zhi
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Wenting Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Tingting Li
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Jinxin Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
10
|
Fusse EJ, Scarante FF, Vicente MA, Marrubia MM, Turcato F, Scomparin DS, Ribeiro MA, Figueiredo MJ, Brigante TAV, Guimarães FS, Campos AC. Anxiogenic doses of rapamycin prevent URB597-induced anti-stress effects in socially defeated mice. Neurosci Lett 2024; 818:137519. [PMID: 37852528 DOI: 10.1016/j.neulet.2023.137519] [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: 08/04/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Repeated exposure to psychosocial stress modulates the endocannabinoid system, particularly anandamide (AEA) signaling in brain regions associated with emotional distress. The mTOR protein regulates various neuroplastic processes in the brain disrupted by stress, including adult hippocampal neurogenesis. This kinase has been implicated in multiple effects of cannabinoid drugs and the anti-stress behavioral effects of psychoactive drugs. Therefore, our hypothesis is that enhancing AEA signaling via pharmacological inhibition of the fatty acid amide hydrolase (FAAH) enzyme induces an anti-stress behavioral effect through an mTOR-dependent mechanism. To test this hypothesis, male C57Bl6 mice were exposed to social defeat stress (SDS) for 7 days and received daily treatment with either vehicle or different doses of the FAAH inhibitor, URB597 (0.1; 0.3; 1 mg/Kg), alone or combined with rapamycin. The results suggested that URB597 induced an inverted U-shaped dose-response curve in mice subjected to SDS (with the intermediate dose of 0.3 mg/kg being anxiolytic, and the higher tested dose of 1 mg/Kg being anxiogenic). In a second independent experiment, rapamycin treatment induced an anxiogenic-like response in control mice. However, in the presence of rapamycin, the anxiolytic dose of URB597 treatment failed to reduce stress-induced anxiety behaviors in mice. SDS exposure altered the hippocampal expression of the mTOR scaffold protein Raptor. Furthermore, the anxiogenic dose of URB597 decreased the absolute number of migrating doublecortin (DCX)-positive cells in the dentate gyrus, suggesting an anti-anxiety effect independent of newly generated/immature neurons. Therefore, our results indicate that in mice exposed to repeated psychosocial stress, URB597 fails to counteract the anxiogenic-like response induced by the pharmacological dampening of mTOR signaling.
Collapse
Affiliation(s)
- Eduardo J Fusse
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Franciele F Scarante
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Maria A Vicente
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Mariana M Marrubia
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Flávia Turcato
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, USA
| | - Davi S Scomparin
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Melissa A Ribeiro
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Maria J Figueiredo
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Tamires A V Brigante
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil
| | - Alline C Campos
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Ave, Ribeirão Preto, Brazil.
| |
Collapse
|
11
|
Jiang M, Huizenga MCW, Wirt JL, Paloczi J, Amedi A, van den Berg RJBHN, Benz J, Collin L, Deng H, Di X, Driever WF, Florea BI, Grether U, Janssen APA, Hankemeier T, Heitman LH, Lam TW, Mohr F, Pavlovic A, Ruf I, van den Hurk H, Stevens AF, van der Vliet D, van der Wel T, Wittwer MB, van Boeckel CAA, Pacher P, Hohmann AG, van der Stelt M. A monoacylglycerol lipase inhibitor showing therapeutic efficacy in mice without central side effects or dependence. Nat Commun 2023; 14:8039. [PMID: 38052772 PMCID: PMC10698032 DOI: 10.1038/s41467-023-43606-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
Monoacylglycerol lipase (MAGL) regulates endocannabinoid 2-arachidonoylglycerol (2-AG) and eicosanoid signalling. MAGL inhibition provides therapeutic opportunities but clinical potential is limited by central nervous system (CNS)-mediated side effects. Here, we report the discovery of LEI-515, a peripherally restricted, reversible MAGL inhibitor, using high throughput screening and a medicinal chemistry programme. LEI-515 increased 2-AG levels in peripheral organs, but not mouse brain. LEI-515 attenuated liver necrosis, oxidative stress and inflammation in a CCl4-induced acute liver injury model. LEI-515 suppressed chemotherapy-induced neuropathic nociception in mice without inducing cardinal signs of CB1 activation. Antinociceptive efficacy of LEI-515 was blocked by CB2, but not CB1, antagonists. The CB1 antagonist rimonabant precipitated signs of physical dependence in mice treated chronically with a global MAGL inhibitor (JZL184), and an orthosteric cannabinoid agonist (WIN55,212-2), but not with LEI-515. Our data support targeting peripheral MAGL as a promising therapeutic strategy for developing safe and effective anti-inflammatory and analgesic agents.
Collapse
Affiliation(s)
- Ming Jiang
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands
| | - Mirjam C W Huizenga
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands
| | - Jonah L Wirt
- Department of Psychological and Brain Sciences, Program in Neuroscience, Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - Janos Paloczi
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute of Health/NIAAA, Rockville, MD, USA
| | - Avand Amedi
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands
| | | | - Joerg Benz
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Ludovic Collin
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Hui Deng
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands
| | - Xinyu Di
- Metabolomics and analytics center, Leiden University, Leiden, Netherlands
| | - Wouter F Driever
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands
| | - Bogdan I Florea
- Department of Bio-organic Synthesis, Leiden University, Leiden, Netherlands
| | - Uwe Grether
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Antonius P A Janssen
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands
| | - Thomas Hankemeier
- Metabolomics and analytics center, Leiden University, Leiden, Netherlands
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden University & Oncode Institute, Leiden, Netherlands
| | | | - Florian Mohr
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands
| | - Anto Pavlovic
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Iris Ruf
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | | - Anna F Stevens
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands
| | - Daan van der Vliet
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands
| | - Tom van der Wel
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands
| | - Matthias B Wittwer
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute of Health/NIAAA, Rockville, MD, USA
| | - Andrea G Hohmann
- Department of Psychological and Brain Sciences, Program in Neuroscience, Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA.
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden University & Oncode Institute, Leiden, Netherlands.
| |
Collapse
|
12
|
The association between adverse childhood experiences and peripartal pain experience. Pain 2023:00006396-990000000-00255. [PMID: 36787580 DOI: 10.1097/j.pain.0000000000002870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/03/2023] [Indexed: 02/16/2023]
Abstract
ABSTRACT Adverse childhood experiences (ACEs) are associated with altered ongoing and evoked pain experiences, which have scarcely been studied for the peripartum period. We aimed to investigate how ACEs affect pain experience in pregnancy and labor. For this noninterventional trial with a short-term follow-up, pregnant women were divided into a trauma group (TG) with ACEs (n = 84) and a control group (CG) without ACEs (n = 107) according to the Childhood Trauma Questionnaire. Pain experience in pregnancy and labor was recorded by self-report and the German Pain Perception Scale. Pain sensitivity prepartum and postpartum was assessed by Quantitative Sensory Testing and a paradigm of conditioned pain modulation (CPM), using pressure pain thresholds (PPTs) and a cold pressor test. The TG showed higher affective and sensory scores for back pain and a more than doubled prevalence of preexisting back pain. Pelvic pain differences were nonsignificant. The TG also exhibited increased affective scores (1.71 ± 0.15 vs 1.33 ± 0.11), but not sensory scores for labor pain during spontaneous delivery. There were no group differences in prepartum pain sensitivity. While PPTs increased through delivery in the CG (clinical CPM), and this PPT change was positively correlated with the experimental CPM (r = 0.55), this was not the case in the TG. The association of ACEs with increased peripartal pain affect and heightened risk for preexisting back pain suggest that such women deserve special care. The dissociation of impaired clinical CPM in women with ACEs and normal prepartum experimental CPM implies at least partly different mechanisms of these 2 manifestations of endogenous pain controls.
Collapse
|
13
|
Bella A, Diego AM, Finn DP, Roche M. Stress-induced changes in nociceptive responding post-surgery in preclinical rodent models. FRONTIERS IN PAIN RESEARCH (LAUSANNE, SWITZERLAND) 2023; 3:1106143. [PMID: 36703943 PMCID: PMC9871907 DOI: 10.3389/fpain.2022.1106143] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023]
Abstract
Chronic post-surgical pain affects up to 85% of individuals depending on the type of surgery, the extent of inflammation, tissue and/or nerve damage. Pre-surgical stress is associated with greater pain intensity, prolonged recovery and is one of the main risk factors for the development of chronic post-surgical pain. Clinically valid animal models provide an important means of examining the mechanisms underlying the effects of stress on post-surgical pain and identifying potential novel therapeutic targets. This review discusses the current data from preclinical animal studies examining the effect of stress on post-surgical pain, the potential underlying mechanisms and gaps in the knowledge that require further investigation.
Collapse
Affiliation(s)
- Ariadni Bella
- Physiology, School of Medicine, University of Galway, Galway, Ireland,Centre for Pain Research, University of Galway, Galway, Ireland,Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Alba M. Diego
- Centre for Pain Research, University of Galway, Galway, Ireland,Galway Neuroscience Centre, University of Galway, Galway, Ireland,Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - David P. Finn
- Centre for Pain Research, University of Galway, Galway, Ireland,Galway Neuroscience Centre, University of Galway, Galway, Ireland,Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Michelle Roche
- Physiology, School of Medicine, University of Galway, Galway, Ireland,Centre for Pain Research, University of Galway, Galway, Ireland,Galway Neuroscience Centre, University of Galway, Galway, Ireland,Correspondence: Michelle Roche
| |
Collapse
|
14
|
Kędziora M, Boccella S, Marabese I, Mlost J, Infantino R, Maione S, Starowicz K. Inhibition of anandamide breakdown reduces pain and restores LTP and monoamine levels in the rat hippocampus via the CB 1 receptor following osteoarthritis. Neuropharmacology 2023; 222:109304. [PMID: 36341807 DOI: 10.1016/j.neuropharm.2022.109304] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/15/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
Abstract
Chronic pain is a persistent, complex condition that contributes to impaired mood, anxiety and emotional problems. Osteoarthritis (OA) is one of the major causes of chronic pain in adults and elderly people. A substantial body of evidence demonstrate that hippocampal neural circuits, especially monoamine dopamine and serotonin levels, contributes to negative affect and avoidance motivation experienced during pain. Current pharmacological strategies for OA patients are unsatisfying and the endocannabinoid system modulation might represent an alternative for the treatment of OA-related pain. In the present study, we used a rat model of osteoarthritis induced by intra-articular injection of sodium monoiodoacetate to assess, 28 days post-induction, the contribution of endocannabinoid system on the possible alteration in pain perception and affective behavior, in LTP and monoamine levels in the lateral entorhinal cortex-dentate gyrus pathway. The results show that OA-related chronic pain induces working memory impairment and depressive-like behavior appearance, diminishes LTP, decreases dopamine levels and increases serotonin levels in the rat dentate gyrus. URB597 administration (i.p., 1 mg/kg) reduces hyperalgesia and mechanical allodynia, improves recognition memory and depressive-live behavior, restores LTP and normalizes monoamine levels in the hippocampus. The effect was observed 60-120 min post-treatment and was blocked by AM251, which proves the action of URB597 via the CB1 receptor. Therefore, our study confirms the role of anandamide in OA-related chronic pain management at the behavioral and hippocampal levels. This article is part of the Special Issue on 'Advances in mechanisms and therapeutic targets relevant to pain'.
Collapse
Affiliation(s)
- Marta Kędziora
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Cracow, Poland
| | - Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Jakub Mlost
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Cracow, Poland
| | - Rosmara Infantino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy; IRCSS, Neuromed, Pozzilli (IS), 86077, Italy; ERG, Endocannabinoid Research Group, CNR, Pozzuoli, Italy
| | - Katarzyna Starowicz
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Cracow, Poland.
| |
Collapse
|
15
|
Schwitter C, Lutz B, Bindila L. Extraction and Simultaneous Quantification of Endocannabinoids and Endocannabinoid-Like Lipids in Biological Tissues. Methods Mol Biol 2023; 2576:9-19. [PMID: 36152174 DOI: 10.1007/978-1-0716-2728-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Extraction and quantification of endocannabinoids from biological tissues is essential to unravel their changes under physiological and pathophysiological conditions. We describe here an analytical protocol for the extraction of endocannabinoids, anandamide (archidonoyl ethanolamide, AEA) and 2-arachidonoyl glycerol (2-AG), and endocannabinoid-like lipids such as palmitoyl ethanolamide (PEA) and oleoyl ethanolamide (OEA), as well as arachidonic acid (AA) from biological tissues using liquid-liquid extraction method and simultaneous quantification by liquid chromatography multiple reaction monitoring (LC/MRM).
Collapse
Affiliation(s)
- Claudia Schwitter
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Laura Bindila
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany.
| |
Collapse
|
16
|
Bourke SL, Schlag AK, O'Sullivan SE, Nutt DJ, Finn DP. Cannabinoids and the endocannabinoid system in fibromyalgia: A review of preclinical and clinical research. Pharmacol Ther 2022; 240:108216. [PMID: 35609718 DOI: 10.1016/j.pharmthera.2022.108216] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/03/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022]
Abstract
Characterised by chronic widespread musculoskeletal pain, generalised hyperalgesia, and psychological distress, fibromyalgia (FM) is a significant unmet clinical need. The endogenous cannabinoid system plays an important role in modulating both pain and the stress response. Here, we appraise the evidence, from preclinical and clinical studies, for a role of the endocannabinoid system in FM and the therapeutic potential of targeting the endocannabinoid system. While many animal models have been used to study FM, the reserpine-induced myalgia model has emerged as perhaps the most translatable to the clinical phenotype. Inhibition of fatty acid amide hydrolase (FAAH) has shown promise in preclinical studies, ameliorating pain- and anxiety-related behaviour . Clinically, there is evidence for alterations in the endocannabinoid system in patients with FM, including single nucleotide polymorphisms and increased levels of circulating endocannabinoids and related N-acylethanolamines. Single entity cannabinoids, cannabis, and cannabis-based medicines in patients with FM show promise therapeutically but limitations in methodology and lack of longitudinal studies to assess efficacy and tolerability preclude the current recommendation for their use in patients with FM. Gaps in the literature that warrant further investigation are discussed, particularly the need for further development of animal models with high validity for the multifaceted nature of FM, balanced studies to eliminate sex-bias in preclinical research, and ultimately, better translation between preclinical and clinical research.
Collapse
Affiliation(s)
- Stephanie L Bourke
- Pharmacology and Therapeutics, School of Medicine, Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Anne Katrin Schlag
- Drug Science, St. Peters House, Wood Street, London, UK; Faculty of Medicine, Department of Brain Sciences, Imperial College London, UK
| | | | - David J Nutt
- Drug Science, St. Peters House, Wood Street, London, UK; Faculty of Medicine, Department of Brain Sciences, Imperial College London, UK
| | - David P Finn
- Pharmacology and Therapeutics, School of Medicine, Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland.
| |
Collapse
|
17
|
Lookfong NA, Raup-Konsavage WM, Silberman Y. Potential Utility of Cannabidiol in Stress-Related Disorders. Cannabis Cannabinoid Res 2022; 8:230-240. [PMID: 36409719 PMCID: PMC10061337 DOI: 10.1089/can.2022.0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background: The endocannabinoid (eCB) system plays an important role in homeostatic regulation of anxiety and stress responses; however, the eCB system can be disrupted following traumatic stressors. Additionally, traumatic or chronic stressors that occur during adulthood or early life can cause long-lasting disturbances in the eCB system. These alterations interfere with hypothalamic-pituitary-adrenal axis function and may be involved in lifelong increased fear and anxiety behaviors as well as increased risk for development of post-traumatic stress disorder (PTSD). Methods: This review focuses on the implications of trauma and significant stressors on eCB functionality and neural pathways, both in adolescence and into adulthood, as well as the current state of testing for CBD efficacy in treating pediatric and adult patients suffering from stress-induced eCB dysregulation. Articles were searched via Pubmed and included studies examining eCB modulation of stress-related disorders in both clinical settings and preclinical models. Conclusion: Given the potential for lifelong alterations in eCB signaling that can mediate stress responsiveness, consideration of pharmaceutical or nutraceutical agents that impact eCB targets may improve clinical outcomes in stress-related disorders. However, caution may be warranted in utilization of medicinal cannabinoid products that contain delta-9-tetrahydrocannabinol due to pronounced euphorigenic effects and potential to exacerbate stress-related behaviors. Other cannabinoid products, such as cannabidiol (CBD), have shown promise in reducing stress-related behaviors in pre-clinical models. Overall, pre-clinical evidence supports CBD as a potential treatment for stress or anxiety disorders resulting from previously stressful events, particularly by reducing fearful behavior and promoting extinction of contextual fear memories, which are hallmarks of PTSD. However, very limited clinical research has been conducted examining the potential effectiveness of CBD in this regard and should be examined further.
Collapse
Affiliation(s)
- Nicole A. Lookfong
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | | | - Yuval Silberman
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
18
|
Morgan A, Adank D, Johnson K, Butler E, Patel S. 2-Arachidonoylglycerol-mediated endocannabinoid signaling modulates mechanical hypersensitivity associated with alcohol withdrawal in mice. Alcohol Clin Exp Res 2022; 46:2010-2024. [PMID: 36125319 PMCID: PMC10091740 DOI: 10.1111/acer.14949] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/05/2022] [Accepted: 09/15/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Alcohol use disorder (AUD) commonly occurs in patients with chronic pain, and a major barrier to achieving abstinence and preventing relapse is the emergence of hyperalgesia during alcohol withdrawal. Elucidating novel therapeutic approaches to target hyperalgesia associated with alcohol withdrawal could have important implications for treating AUD. Here, we examined the role of 2-arachidonoylglycerol (2-AG)-mediated endocannabinoid (eCB) signaling in the regulation of hyperalgesia associated with alcohol withdrawal in mice. We tested the hypothesis that pharmacological augmentation of 2-AG signaling could reduce hyperalgesia during withdrawal. METHODS Male and female C57BL/6J mice were tested during withdrawal from a continuous access two-bottle choice (2BC) paradigm to investigate how eCB signaling modulates mechanical and thermal sensitivity during withdrawal. Mice were pretreated with the monoacylglycerol lipase (MAGL) inhibitor JZL184 to elevate levels of 2-AG. Rimonabant or AM630 were given to block CB1 and CB2 receptor activity, respectively. DO34 was given to reduce 2-AG by inhibiting the 2-AG synthetic enzyme diacylglycerol lipase (DAGL). RESULTS After 72 h of withdrawal, male and female mice exhibited increased mechanical, but not thermal, hypersensitivity, which normalized by 7 days. This effect was reversed by pretreatment with JZL184. The effects of JZL184 were prevented by coadministration of either the CB1 or the CB2 antagonist. DO34, Rimonabant, and AM630 exacerbated mechanical hypersensitivity during alcohol withdrawal, causing an earlier onset and persistent hypersensitivity even 1 week into withdrawal. CONCLUSIONS Our findings demonstrate the critical role of 2-AG signaling in the bidirectional regulation of mechanical sensitivity during alcohol withdrawal, with enhancement of 2-AG levels reducing sensitivity, and inhibition of 2-AG signaling exacerbating sensitivity. These data suggest that 2-AG augmentation represents a novel approach to the treatment of alcohol withdrawal-associated hyperalgesia and AUD in patients with comorbid pain disorders.
Collapse
Affiliation(s)
- Amanda Morgan
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Danielle Adank
- Vanderbilt Brain Institute, Vanderbilt UniversityNashvilleTennesseeUSA
| | - Keenan Johnson
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Emily Butler
- Interdisciplinary Program in NeuroscienceVanderbilt UniversityNashvilleTennesseeUSA
| | - Sachin Patel
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| |
Collapse
|
19
|
Persistent muscle hyperalgesia after adolescent stress is exacerbated by a mild-nociceptive input in adulthood and is associated with microglia activation. Sci Rep 2022; 12:18324. [PMID: 36316425 PMCID: PMC9622712 DOI: 10.1038/s41598-022-21808-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
Abstract
Non-specific low back pain (LBP) is a major global disease burden and childhood adversity predisposes to its development. The mechanisms are largely unknown. Here, we investigated if adversity in young rats augments mechanical hyperalgesia and how spinal cord microglia contribute to this. Adolescent rats underwent restraint stress, control animals were handled. In adulthood, all rats received two intramuscular injections of NGF/saline or both into the lumbar multifidus muscle. Stress induced in rats at adolescence lowered low back pressure pain threshold (PPT; p = 0.0001) and paw withdrawal threshold (PWT; p = 0.0007). The lowered muscle PPT persisted throughout adulthood (p = 0.012). A subsequent NGF in adulthood lowered only PPT (d = 0.87). Immunohistochemistry revealed changes in microglia morphology: stress followed by NGF induced a significant increase in ameboid state (p < 0.05). Repeated NGF injections without stress showed significantly increased cell size in surveilling and bushy states (p < 0.05). Thus, stress in adolescence induced persistent muscle hyperalgesia that can be enhanced by a mild-nociceptive input. The accompanying morphological changes in microglia differ between priming by adolescent stress and by nociceptive inputs. This novel rodent model shows that adolescent stress is a risk factor for the development of LBP in adulthood and that morphological changes in microglia are signs of spinal mechanisms involved.
Collapse
|
20
|
Ferrarini EG, Paes RS, Baldasso GM, de Assis PM, Gouvêa MC, Cicco PD, Raposo NRB, Capasso R, Moreira ELG, Dutra RC. Broad-spectrum cannabis oil ameliorates reserpine-induced fibromyalgia model in mice. Biomed Pharmacother 2022; 154:113552. [PMID: 35988425 DOI: 10.1016/j.biopha.2022.113552] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/28/2022] [Accepted: 08/14/2022] [Indexed: 01/25/2023] Open
Abstract
Fibromyalgia (FM) is an idiopathic disorder characterized by generalized pain and associated symptoms such as depression and anxiety. Cannabis sativa shows different pharmacological activities, such as analgesic, anti-inflammatory, neuroprotective, and immunomodulatory. Associated with this, the use of an oil with low concentrations of THC can reduce the psychomimetic adverse effects of the plant. Therefore, the present study aimed to evaluate the analgesic effect of broad-spectrum cannabis oil with low THC concentration in an experimental model of FM. Mechanical hyperalgesia, thermal allodynia, depressive- and anxious-related behavior, and locomotor activity were evaluated after reserpine (0.25 mg/kg; injected subcutaneously (s.c.) once daily for three consecutive days) administration. Our results showed that oral administration of broad-spectrum cannabis oil (0.1, 1, and 3 mg/kg, p.o.) in a single dose on the 4th day inhibited mechanical hyperalgesia and thermal allodynia induced by reserpine. Relevantly, treatment during four days with broad-spectrum cannabis oil (0.1 mg/kg, p.o.) reduced mechanical hyperalgesia 1 h after reserpine administration. Intraplantar treatment with cannabis oil significantly reversed mechanical and heat thermal nociception induced by reserpine injection. Interestingly, spinal and supraspinal administration of broad-spectrum cannabis oil completely inhibited mechanical hyperalgesia and thermal sensitivity induced by reserpine. The repeated cannabis oil administration, given daily for 14 days, markedly mitigated the mechanical and thermal sensitivity during the FM model, and its reduced depressive-like behavior induced by reserpine. In summary, broad-spectrum cannabis oil is an effective alternative to reverse the reserpine-induced fibromyalgia model.
Collapse
Affiliation(s)
- Eduarda Gomes Ferrarini
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil; Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil.
| | - Rodrigo Sebben Paes
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil.
| | - Gabriela Mantovani Baldasso
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil.
| | - Pollyana Mendonça de Assis
- Center of Research and Innovation in Health Sciences, School of Pharmacy, Universidade Federal de Juiz de Fora, 36036-330 Juiz de Fora, MG, Brazil.
| | - Murilo Chaves Gouvêa
- Associação Brasileira de Apoio Cannabis e Esperança, Parque Sólon de Lucena, 697, 58028-470 João Pessoa, PB, Brazil.
| | - Paola De Cicco
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy.
| | - Nádia Rezende Barbosa Raposo
- Center of Research and Innovation in Health Sciences, School of Pharmacy, Universidade Federal de Juiz de Fora, 36036-330 Juiz de Fora, MG, Brazil.
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy.
| | - Eduardo Luiz Gasnhar Moreira
- Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil.
| | - Rafael Cypriano Dutra
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil; Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil.
| |
Collapse
|
21
|
The antidepressant and anxiolytic effects of cannabinoids in chronic unpredictable stress: a preclinical systematic review and meta-analysis. Transl Psychiatry 2022; 12:217. [PMID: 35641487 PMCID: PMC9156762 DOI: 10.1038/s41398-022-01967-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 04/22/2022] [Accepted: 05/04/2022] [Indexed: 12/09/2022] Open
Abstract
Neuroscience research presents contradictory evidence in support of both the protective and destructive effects of cannabinoids in depression. Therefore, this systematic review and meta-analysis summarizes the existing preclinical literature on the effects of cannabinoid administration in the chronic unpredictable stress model of depression in order to evaluate the effects of cannabinoids and identify gaps in the literature. After protocol registration (PROSPERO #CRD42020219986), we systematically searched Scopus, Embase, Psychology & Behavioral Sciences Collection, APA PsychINFO, PubMed, CINAHL Complete, and ProQuest Dissertations & Theses Global from the earliest record of the databases, February 1964, to November 2020 for articles that met inclusion criteria (e.g., rodent subjects and administration of a cannabinoid. A total of 26 articles were included representing a sample size estimate of 1132 rodents with the majority of articles administering daily intraperitoneal injections during chronic unpredictable stress. These articles were evaluated using a modified SYRCLE's risk-of-bias tool. For each continuous behavioral measure, the standardized mean difference was calculated between cannabinoid and vehicle groups in rodents subjected to chronic unpredictable stress. The effects of cannabinoids on depressive-like behavior was evaluated using a multilevel mixed-effects model with effect size weights nested within control groups. Cannabinoid administration moderately improved the pooled negative effects of chronic unpredictable stress on anhedonia, learned helplessness, novelty suppressed feeding, time in the anxiogenic context, and entries into the anxiogenic context. Although the interpretations are limited, these findings suggest that with further investigation, cannabinoids may be a viable long-term treatment for stress-related psychopathologies such as depression.
Collapse
|
22
|
Zhang Y, Ke J, Zhou Y, Liu X, Huang T, Wang F. Sex-specific characteristics of cells expressing the cannabinoid 1 receptor in the dorsal horn of the lumbar spinal cord. J Comp Neurol 2022; 530:2451-2473. [PMID: 35580011 DOI: 10.1002/cne.25342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 12/20/2022]
Abstract
It is becoming increasingly clear that robust sex differences exist in the processing of acute and chronic pain in both rodents and humans. However, the underlying mechanism has not been well characterized. The dorsal horn of the lumbar spinal cord is the fundamental building block of ascending and descending pain pathways. It has been shown that numerous neurotransmitter and neuromodulator systems in the spinal cord, including the endocannabinoid system and its main receptor, the cannabinoid 1 receptor (CB1 R), play vital roles in processing nociceptive information. Our previous findings have shown that CB1 R mRNA is widely expressed in the brain in sex-dependent patterns. However, the sex-, lamina-, and cell-type-specific characteristics of CB1 R expression in the spinal cord have not been fully described. In this study, the CB1 R-iCre-EGFP mouse strain was generated to label and identify CB1 R-positive (CB1 RGFP ) cells. We reported no sex difference in CB1 R expression in the lumbar dorsal horn of the spinal cord, but a dynamic distribution within superficial laminae II and III in female mice between estrus and nonestrus phases. Furthermore, the cell-type-specific CB1 R expression pattern in the dorsal horn was similar in both sexes. Over 50% of CB1 RGFP cells were GABAergic neurons, and approximately 25% were glycinergic and 20-30% were glutamatergic neurons. The CB1 R-expressing cells also represented a subset of spinal projection neurons. Overall, our work indicates a highly consistent distribution pattern of CB1 RGFP cells in the dorsal horn of lumbar spinal cord in males and females.
Collapse
Affiliation(s)
- Yulin Zhang
- Shenzhen Key Lab of Translational Research for Brain Diseases, Shenzhen Key Lab of Drug Addiction, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jin Ke
- Shenzhen Key Lab of Translational Research for Brain Diseases, Shenzhen Key Lab of Drug Addiction, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Zhou
- Shenzhen Key Lab of Translational Research for Brain Diseases, Shenzhen Key Lab of Drug Addiction, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xue Liu
- Shenzhen Key Lab of Translational Research for Brain Diseases, Shenzhen Key Lab of Drug Addiction, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Tianwen Huang
- Shenzhen Key Lab of Translational Research for Brain Diseases, Shenzhen Key Lab of Drug Addiction, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Feng Wang
- Shenzhen Key Lab of Translational Research for Brain Diseases, Shenzhen Key Lab of Drug Addiction, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
23
|
Yang X, Guo JY, Jiang YN, Liu MM, Li QY, Li JY, Wei XJ, Wan GH, Shi JL. Valeriana jatamansi Jones ex Roxb. Against Post-Traumatic Stress Disorder, Network Pharmacological Analysis, and In Vivo Evaluation. Front Pharmacol 2021; 12:764548. [PMID: 34950028 PMCID: PMC8688958 DOI: 10.3389/fphar.2021.764548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/11/2021] [Indexed: 12/31/2022] Open
Abstract
Zhi zhu xiang (ZZX) is the root and rhizome of Valeriana jatamansi Jones ex Roxb. Recent studies have shown that ZZX can exert antianxiety, antidepressant, and sedative effects. Because post-traumatic stress disorder (PTSD) is similar to depression and anxiety in terms of its etiology, pathogenesis, and clinical manifestations, it is possible that ZZX may also be useful for the prevention and treatment of PTSD. In this study, a mouse model of PTSD was established and used to study the pharmacological action of a 95% ethanol extract of ZZX on PTSD via a series of classic behavioral tests. We found that a 95% ethanol extract of ZZX was indeed effective for relieving the symptoms of PTSD in mice. Moreover, network pharmacology analysis was used to predict the potential active ingredients, targets, and possible pathways of ZZX in the treatment of PTSD. The neurotransmitter system, the hypothalamic-pituitary-adrenal (HPA) axis, and the endocannabinoid (eCB) system were identified to be the most likely pathways for anti-PTSD action in ZZX. Due to the lack of a falsification mechanism in network pharmacology, in vivo tests were carried out in mice, and the expression levels of neurotransmitters, hormones, and genes of key targets were detected by enzyme-linked immunosorbent assay and real-time PCR to further verify this inference. Analysis showed that the levels of norepinephrine, 5-hydroxytryptamine, and glutamic acid were increased in the hippocampus, prefrontal cortex, and amygdala of PTSD mice, while the levels of dopamine and γ-aminobutyric acid were decreased in these brain regions; furthermore, ZZX could restore the expression of these factors, at least to a certain extent. The levels of adrenocorticotropic hormone, corticosterone, and corticotropin-releasing hormone were increased in these different brain regions and the serum of PTSD mice; these effects could be reversed by ZZX to a certain extent. The expression levels of cannabinoid receptor 1 and diacylglycerol lipase α mRNA were decreased in PTSD mice, while the levels of fatty acid amide hydrolase and monoacylglycerol lipase mRNA were increased; these effects were restored by ZZX to a certain extent. In conclusion, our findings suggest that ZZX may provide new therapeutic pathways for treating PTSD by the regulation of neurotransmitters, the HPA, and expression levels of eCB-related genes in the brain.
Collapse
Affiliation(s)
- Xue Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jian-You Guo
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Ya-Ni Jiang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Meng-Meng Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Qiu-Yu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jia-Yuan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Jia Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guo-Hui Wan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jin-Li Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
24
|
Zanfirescu A, Ungurianu A, Mihai DP, Radulescu D, Nitulescu GM. Targeting Monoacylglycerol Lipase in Pursuit of Therapies for Neurological and Neurodegenerative Diseases. Molecules 2021; 26:5668. [PMID: 34577139 PMCID: PMC8468992 DOI: 10.3390/molecules26185668] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022] Open
Abstract
Neurological and neurodegenerative diseases are debilitating conditions, and frequently lack an effective treatment. Monoacylglycerol lipase (MAGL) is a key enzyme involved in the metabolism of 2-AG (2-arachidonoylglycerol), a neuroprotective endocannabinoid intimately linked to the generation of pro- and anti-inflammatory molecules. Consequently, synthesizing selective MAGL inhibitors has become a focus point in drug design and development. The purpose of this review was to summarize the diverse synthetic scaffolds of MAGL inhibitors concerning their potency, mechanisms of action and potential therapeutic applications, focusing on the results of studies published in the past five years. The main irreversible inhibitors identified were derivatives of hexafluoroisopropyl alcohol carbamates, glycol carbamates, azetidone triazole ureas and benzisothiazolinone, whereas the most promising reversible inhibitors were derivatives of salicylketoxime, piperidine, pyrrolidone and azetidinyl amides. We reviewed the results of in-depth chemical, mechanistic and computational studies on MAGL inhibitors, in addition to the results of in vitro findings concerning selectivity and potency of inhibitors, using the half maximal inhibitory concentration (IC50) as an indicator of their effect on MAGL. Further, for highlighting the potential usefulness of highly selective and effective inhibitors, we examined the preclinical in vivo reports regarding the promising therapeutic applications of MAGL pharmacological inhibition.
Collapse
Affiliation(s)
| | - Anca Ungurianu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (A.Z.); (D.P.M.); (D.R.); (G.M.N.)
| | | | | | | |
Collapse
|
25
|
Sphingolipid and Endocannabinoid Profiles in Adult Attention Deficit Hyperactivity Disorder. Biomedicines 2021; 9:biomedicines9091173. [PMID: 34572359 PMCID: PMC8467584 DOI: 10.3390/biomedicines9091173] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/04/2021] [Accepted: 09/05/2021] [Indexed: 01/27/2023] Open
Abstract
Genes encoding endocannabinoid and sphingolipid metabolism pathways were suggested to contribute to the genetic risk towards attention deficit hyperactivity disorder (ADHD). The present pilot study assessed plasma concentrations of candidate endocannabinoids, sphingolipids and ceramides in individuals with adult ADHD in comparison with healthy controls and patients with affective disorders. Targeted lipid analyses of 23 different lipid species were performed in 71 mental disorder patients and 98 healthy controls (HC). The patients were diagnosed with adult ADHD (n = 12), affective disorder (major depression, MD n = 16 or bipolar disorder, BD n = 6) or adult ADHD with comorbid affective disorders (n = 37). Canonical discriminant analysis and CHAID analyses were used to identify major components that predicted the diagnostic group. ADHD patients had increased plasma concentrations of sphingosine-1-phosphate (S1P d18:1) and sphinganine-1-phosphate (S1P d18:0). In addition, the endocannabinoids, anandamide (AEA) and arachidonoylglycerol were increased. MD/BD patients had increased long chain ceramides, most prominently Cer22:0, but low endocannabinoids in contrast to ADHD patients. Patients with ADHD and comorbid affective disorders displayed increased S1P d18:1 and increased Cer22:0, but the individual lipid levels were lower than in the non-comorbid disorders. Sphingolipid profiles differ between patients suffering from ADHD and affective disorders, with overlapping patterns in comorbid patients. The S1P d18:1 to Cer22:0 ratio may constitute a diagnostic or prognostic tool.
Collapse
|
26
|
Le J, Peng R, Li Y. Trimethylamine-N-oxide and Precursors as Novel Potential Biomarkers for Anxiety Disorder. Lab Med 2021; 53:177-182. [PMID: 34480186 DOI: 10.1093/labmed/lmab063] [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] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Anxiety disorder (AD) is closely related to changes in the composition of the gut microbiota, and changes in gut microbiota abundance affect the synthesis of trimethylamine N-oxide (TMAO). OBJECTIVE To explore whether TMAO is related to and serves as a potential biomarker for the diagnosis of AD. METHODS The concentrations of TMAO, choline, and betaine were analyzed in 60 patients with AD and 60 control individuals using a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. RESULTS Compared with controls, TMAO was significantly reduced in patients with AD. Logistic regression analysis demonstrated that decreased TMAO concentrations were associated with an increased risk of AD. The multiplication of TMAO and its precursors (choline and betaine) produced the best AUC for the diagnosis of AD (AUC = 0.847; 95% CI, 0.780-0.914; P <.001). CONCLUSION The decrease of TMAO concentration is related to the increase of anxiety disorder. TMAO and precursors could be identified as novel potential biomarkers for the diagnosis of AD.
Collapse
Affiliation(s)
- Juan Le
- Clinical Laboratory, Renmin Hospital, Wuhan University, Wuhan, China
| | - Rui Peng
- Clinical Laboratory, Renmin Hospital, Wuhan University, Wuhan, China
| | - Yan Li
- Clinical Laboratory, Renmin Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
27
|
Xu X, Wu K, Ma X, Wang W, Wang H, Huang M, Luo L, Su C, Yuan T, Shi H, Han J, Wang A, Xu T. mGluR5-Mediated eCB Signaling in the Nucleus Accumbens Controls Vulnerability to Depressive-Like Behaviors and Pain After Chronic Social Defeat Stress. Mol Neurobiol 2021; 58:4944-4958. [PMID: 34227060 DOI: 10.1007/s12035-021-02469-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022]
Abstract
Stress contributes to major depressive disorder (MDD) and chronic pain, which affect a significant portion of the global population, but researchers have not clearly determined how these conditions are initiated or amplified by stress. The chronic social defeat stress (CSDS) model is a mouse model of psychosocial stress that exhibits depressive-like behavior and chronic pain. We hypothesized that metabotropic glutamate receptor 5 (mGluR5) expressed in the nucleus accumbens (NAc) normalizes the depressive-like behaviors and pain following CSDS. Here, we show that CSDS induced both pain and social avoidance and that the level of mGluR5 decreased in susceptible mice. Overexpression of mGluR5 in the NAc shell and core prevented the development of depressive-like behaviors and pain in susceptible mice, respectively. Conversely, depression-like behaviors and pain were exacerbated in mice with mGluR5 knockdown in the NAc shell and core, respectively, compared to control mice subjected to 3 days of social defeat stress. Furthermore, (RS)-2-chloro-5-hydroxyphenylglycine (CHPG), an mGluR5 agonist, reversed the reduction in the level of the endocannabinoid (eCB) 2-arachidonoylglycerol (2-AG) in the NAc of susceptible mice, an effect that was blocked by 3-((2-methyl-1, 3-thiazol-4-yl) ethynyl) pyridine hydrochloride (MTEP), an mGluR5 antagonist. In addition, the injection of CHPG into the NAc shell and core normalized depressive-like behaviors and pain, respectively, and these effects were inhibited by AM251, a cannabinoid type 1 receptor (CB1R) antagonist. Based on these results, mGluR5-mediated eCB production in the NAc relieves stress-induced depressive-like behaviors and pain.
Collapse
Affiliation(s)
- Xiaotao Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Kaixuan Wu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Xiaqing Ma
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Wenying Wang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Haiyan Wang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Min Huang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Limin Luo
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Chen Su
- Department of Anesthesiology and Pain Medicine, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, People's Republic of China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China
| | - Haibo Shi
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, 200233, People's Republic of China
| | - Ji Han
- Internal medicine of TCM, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, No.164 Lanxi Road, Shanghai, 200062, China.
| | - Aizhong Wang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| | - Tao Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China. .,Department of Anesthesiology, Tongzhou People's Hospital, Nantong, 226300, China.
| |
Collapse
|
28
|
Petrie GN, Nastase AS, Aukema RJ, Hill MN. Endocannabinoids, cannabinoids and the regulation of anxiety. Neuropharmacology 2021; 195:108626. [PMID: 34116110 DOI: 10.1016/j.neuropharm.2021.108626] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/11/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022]
Abstract
Cannabis has been used for hundreds of years, with its ability to dampen feelings of anxiety often reported as a primary reason for use. Only recently has the specific role cannabinoids play in anxiety been thoroughly investigated. Here we discuss the body of evidence describing how endocannabinoids and exogenous cannabinoids are capable of regulating the generation and termination of anxiety states. Disruption of the endogenous cannabinoid (eCB) system following genetic manipulation, pharmacological intervention or stress exposure reliably leads to the generation of an anxiety state. On the other hand, upregulation of eCB signaling is capable of alleviating anxiety-like behaviors in multiple paradigms. When considering exogenous cannabinoid administration, cannabinoid receptor 1 (CB1) agonists have a biphasic, dose-dependent effect on anxiety such that low doses are anxiolytic while high doses are anxiogenic, a phenomenon that is evident in both rodent models and humans. Translational studies investigating a loss of function mutation in the gene for fatty acid amide hydrolase, the enzyme responsible for metabolizing AEA, have also shown that AEA signaling regulates anxiety in humans. Taken together, evidence reviewed here has outlined a convincing argument for cannabinoids being powerful regulators of both the manifestation and amelioration of anxiety symptoms, and highlights the therapeutic potential of targeting the eCB system for the development of novel classes of anxiolytics. This article is part of the special issue on 'Cannabinoids'.
Collapse
Affiliation(s)
- Gavin N Petrie
- Hotchkiss Brain Institute and the Mathison Centre for Mental Health Education and Research, Departments of Cell Biology and Anatomy & Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Andrei S Nastase
- Hotchkiss Brain Institute and the Mathison Centre for Mental Health Education and Research, Departments of Cell Biology and Anatomy & Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Robert J Aukema
- Hotchkiss Brain Institute and the Mathison Centre for Mental Health Education and Research, Departments of Cell Biology and Anatomy & Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute and the Mathison Centre for Mental Health Education and Research, Departments of Cell Biology and Anatomy & Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
29
|
Ahmed I, Rehman SU, Shahmohamadnejad S, Zia MA, Ahmad M, Saeed MM, Akram Z, Iqbal HMN, Liu Q. Therapeutic Attributes of Endocannabinoid System against Neuro-Inflammatory Autoimmune Disorders. Molecules 2021; 26:3389. [PMID: 34205169 PMCID: PMC8199938 DOI: 10.3390/molecules26113389] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/11/2021] [Accepted: 05/29/2021] [Indexed: 02/05/2023] Open
Abstract
In humans, various sites like cannabinoid receptors (CBR) having a binding affinity with cannabinoids are distributed on the surface of different cell types, where endocannabinoids (ECs) and derivatives of fatty acid can bind. The binding of these substance(s) triggers the activation of specific receptors required for various physiological functions, including pain sensation, memory, and appetite. The ECs and CBR perform multiple functions via the cannabinoid receptor 1 (CB1); cannabinoid receptor 2 (CB2), having a key effect in restraining neurotransmitters and the arrangement of cytokines. The role of cannabinoids in the immune system is illustrated because of their immunosuppressive characteristics. These characteristics include inhibition of leucocyte proliferation, T cells apoptosis, and induction of macrophages along with reduced pro-inflammatory cytokines secretion. The review seeks to discuss the functional relationship between the endocannabinoid system (ECS) and anti-tumor characteristics of cannabinoids in various cancers. The therapeutic potential of cannabinoids for cancer-both in vivo and in vitro clinical trials-has also been highlighted and reported to be effective in mice models in arthritis for the inflammation reduction, neuropathic pain, positive effect in multiple sclerosis and type-1 diabetes mellitus, and found beneficial for treating in various cancers. In human models, such studies are limited; thereby, further research is indispensable in this field to get a conclusive outcome. Therefore, in autoimmune disorders, therapeutic cannabinoids can serve as promising immunosuppressive and anti-fibrotic agents.
Collapse
Affiliation(s)
- Ishtiaq Ahmed
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530005, China;
- School of Medical Science, Gold Coast Campus, Griffith University, Southport, QLD 4222, Australia;
| | - Saif Ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530005, China;
| | - Shiva Shahmohamadnejad
- Department of Clinical Biochemistry, School of medicine, Tehran University of Medical Sciences, Tehran 14176-13151, Iran;
| | - Muhammad Anjum Zia
- Enzyme Biotechnology Laboratory, Department of Biochemistry, University of Agriculture, Faisalabad 38040, Pakistan; (M.A.Z.); (M.M.S.)
| | - Muhammad Ahmad
- Faculty of Veterinary Sciences, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences (SBBUVAS), Sakrand 67210, Pakistan;
| | - Muhammad Muzammal Saeed
- Enzyme Biotechnology Laboratory, Department of Biochemistry, University of Agriculture, Faisalabad 38040, Pakistan; (M.A.Z.); (M.M.S.)
| | - Zain Akram
- School of Medical Science, Gold Coast Campus, Griffith University, Southport, QLD 4222, Australia;
| | - Hafiz M. N. Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, 64849 Monterrey, Mexico;
| | - Qingyou Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530005, China;
| |
Collapse
|
30
|
Silveira KM, Wegener G, Joca SRL. Targeting 2-arachidonoylglycerol signalling in the neurobiology and treatment of depression. Basic Clin Pharmacol Toxicol 2021; 129:3-14. [PMID: 33905617 DOI: 10.1111/bcpt.13595] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/14/2021] [Accepted: 04/22/2021] [Indexed: 12/22/2022]
Abstract
The endocannabinoid 2-arachidonoylglycerol (2-AG) is an atypical neurotransmitter synthesized on demand in response to a wide range of stimuli, including exposure to stress. Through the activation of cannabinoid receptors, 2-AG can interfere with excitatory and inhibitory neurotransmission in different brain regions and modulate behavioural, endocrine and emotional components of the stress response. Exposure to chronic or intense unpredictable stress predisposes to maladaptive behaviour and is one of the main risk factors involved in developing mood disorders, such as major depressive disorder (MDD). In this review, we describe the molecular mechanisms involved in 2-AG signalling in the brain of healthy and stressed animals and discuss how such mechanisms could modulate stress adaptation and susceptibility to depression. Furthermore, we review preclinical evidence indicating that the pharmacological modulation of 2-AG signalling stands as a potential new therapeutic target in treating MDD. Particular emphasis is given to the pharmacological augmentation of 2-AG levels by monoacylglycerol lipase (MAGL) inhibitors and the modulation of CB2 receptors.
Collapse
Affiliation(s)
- Kennia M Silveira
- School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of Sao Paulo (USP), Ribeirao Preto, Brazil.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sâmia R L Joca
- School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of Sao Paulo (USP), Ribeirao Preto, Brazil.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
31
|
Qin Z, Zhang L, Zasloff MA, Stewart AFR, Chen HH. Ketamine's schizophrenia-like effects are prevented by targeting PTP1B. Neurobiol Dis 2021; 155:105397. [PMID: 34015491 DOI: 10.1016/j.nbd.2021.105397] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/18/2021] [Accepted: 05/14/2021] [Indexed: 11/26/2022] Open
Abstract
Subanesthetic doses of ketamine induce schizophrenia-like behaviors in mice including hyperlocomotion and deficits in working memory and sensorimotor gating. Here, we examined the effect of in vivo ketamine administration on neuronal properties and endocannabinoid (eCB)-dependent modulation of synaptic transmission onto layer 2/3 pyramidal neurons in brain slices of the prefrontal cortex, a region tied to the schizophrenia-like behavioral phenotypes of ketamine. Since deficits in working memory and sensorimotor gating are tied to activation of the tyrosine phosphatase PTP1B in glutamatergic neurons, we asked whether PTP1B contributes to these effects of ketamine. Ketamine increased membrane resistance and excitability of pyramidal neurons. Systemic pharmacological inhibition of PTP1B by Trodusquemine restored these neuronal properties and prevented each of the three main ketamine-induced behavior deficits. Ketamine also reduced mobilization of eCB by pyramidal neurons, while unexpectedly reducing their inhibitory inputs, and these effects of ketamine were blocked or occluded by PTP1B ablation in glutamatergic neurons. While ablation of PTP1B in glutamatergic neurons prevented ketamine-induced deficits in memory and sensorimotor gating, it failed to prevent hyperlocomotion (a psychosis-like phenotype). Taken together, these results suggest that PTP1B in glutamatergic neurons mediates ketamine-induced deficits in eCB mobilization, memory and sensorimotor gating whereas PTP1B in other cell types contributes to hyperlocomotion. Our study suggests that the PTP1B inhibitor Trodusquemine may represent a new class of fast-acting antipsychotic drugs to treat schizophrenia-like symptoms.
Collapse
Affiliation(s)
- Zhaohong Qin
- Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada
| | - Li Zhang
- Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada
| | - Michael A Zasloff
- Georgetown University School of Medicine, MedStar Georgetown Transplant Institute, Washington D.C. 2007, USA
| | - Alexandre F R Stewart
- University of Ottawa Heart Institute, Ottawa, ON K1Y4W7, Canada; Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| | - Hsiao-Huei Chen
- Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Brain and Mind Institute, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
32
|
Rat dorsal horn neurons primed by stress develop a long-lasting manifest sensitization after a short-lasting nociceptive low back input. Pain Rep 2021; 6:e904. [PMID: 33688602 PMCID: PMC7935483 DOI: 10.1097/pr9.0000000000000904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/29/2020] [Accepted: 01/15/2021] [Indexed: 11/26/2022] Open
Abstract
Background A single injection of nerve growth factor (NGF) into a low back muscle induces a latent sensitization of rat dorsal horn neurons (DHNs) that primes for a manifest sensitization by a subsequent second NGF injection. Repeated restraint stress also causes a latent DHN sensitization. Objective In this study, we investigated whether repeated restraint stress followed by a single NGF injection causes a manifest sensitization of DHNs. Methods Rats were stressed repeatedly in a narrow plastic restrainer (1 hour on 12 consecutive days). Control animals were handled but not restrained. Two days after stress paradigm, behavioral tests and electrophysiological in vivo recordings from single DHNs were performed. Mild nociceptive low back input was induced by a single NGF injection into the lumbar multifidus muscle just before the recording started. Results Restraint stress slightly lowered the low back pressure pain threshold (Cohen d = 0.83). Subsequent NGF injection increased the proportion of neurons responsive to deep low back input (control + NGF: 14%, stress + NGF: 39%; P = 0.041), mostly for neurons with input from outside the low back (7% vs 26%; P = 0.081). There was an increased proportion of neurons with resting activity (28% vs 55%; P = 0.039), especially in neurons having deep input (0% vs 26%; P = 0.004). Conclusions The results indicate that stress followed by a short-lasting nociceptive input causes manifest sensitization of DHNs to deep input, mainly from tissue outside the low back associated with an increased resting activity. These findings on neuronal mechanisms in our rodent model suggest how stress might predispose to radiating pain in patients.
Collapse
|
33
|
Sessler K, Blechschmidt V, Hoheisel U, Mense S, Schirmer L, Treede RD. Spinal cord fractalkine (CX3CL1) signaling is critical for neuronal sensitization in experimental nonspecific, myofascial low back pain. J Neurophysiol 2021; 125:1598-1611. [PMID: 33596743 DOI: 10.1152/jn.00348.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Neuroactive substances released by activated microglia contribute to hyperexcitability of spinal dorsal horn neurons in many animal models of chronic pain. An important feedback loop mechanism is via release of fractalkine (CX3CL1) from primary afferent terminals and dorsal horn neurons and binding to CX3CR1 receptors on microglial cells. We studied the involvement of fractalkine signaling in latent and manifest spinal sensitization induced by two injections of nerve growth factor (NGF) into the lumbar multifidus muscle as a model for myofascial low back pain. Single dorsal horn neurons were recorded in vivo to study their receptive fields and spontaneous activity. Under intrathecal vehicle application, the two NGF injections led to an increased proportion of neurons responding to stimulation of deep tissues (41%), to receptive field expansion into the hindlimb (15%), and to resting activity (53%). Blocking fractalkine signaling by continuous intrathecal administration of neutralizing antibodies completely prevented these signs of spinal sensitization to a similar extent as in a previous study with the microglia inhibitor minocycline. Reversely, fractalkine itself induced similar sensitization in a dose-dependent manner (for 200 ng/mL: 45% deep tissue responses, 24% receptive field expansion, and 45% resting activity) as repeated nociceptive stimulation by intramuscular NGF injections. A subsequent single NGF injection did not have an additive effect. Our data suggest that neuron-to-microglia signaling via the CX3CL1-CX3CR1 pathway is critically involved in the initiation of nonspecific, myofascial low back pain through repetitive nociceptive stimuli.NEW & NOTEWORTHY Blocking fractalkine signaling by neutralizing antibodies completely prevented spinal sensitization induced by repetitive mild nociceptive input [2 nerve growth factor (NGF) injections into the multifidus muscle] Conversely, fractalkine given intrathecally caused the same pattern of spinal sensitization as the nociceptive NGF injections. Fractalkine signaling is critically involved in sensitization of dorsal horn neurons induced by repeated nociceptive low back muscle stimulation and may hence be a potential target for the prevention of nonspecific, myofascial low back pain.
Collapse
Affiliation(s)
- Katharina Sessler
- Department of Neurophysiology, Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Vivian Blechschmidt
- Department of Neurophysiology, Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ulrich Hoheisel
- Department of Neurophysiology, Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Siegfried Mense
- Department of Neurophysiology, Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lucas Schirmer
- Department of Neurology, Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rolf-Detlef Treede
- Department of Neurophysiology, Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
34
|
Bitencourt RM, Takahashi RN, Carlini EA. From an Alternative Medicine to a New Treatment for Refractory Epilepsies: Can Cannabidiol Follow the Same Path to Treat Neuropsychiatric Disorders? Front Psychiatry 2021; 12:638032. [PMID: 33643100 PMCID: PMC7905048 DOI: 10.3389/fpsyt.2021.638032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/12/2021] [Indexed: 12/25/2022] Open
Abstract
Although cannabis has been known for ages as an "alternative medicine" to provide relief from seizures, pain, anxiety, and inflammation, there had always been a limited scientific review to prove and establish its use in clinics. Early studies carried out by Carlini's group in Brazil suggested that cannabidiol (CBD), a non-psychotropic phytocannabinoid present in Cannabis sativa, has anticonvulsant properties in animal models and reduced seizure frequency in limited human trials. Over the past few years, the potential use of cannabis extract in refractory epilepsy, including childhood epilepsies such as Dravet's syndrome and Lennox-Gastaut Syndrome, has opened a new era of treating epileptic patients. Thus, a considerable number of pre-clinical and clinical studies have provided strong evidence that phytocannabinoids has anticonvulsant properties, as well as being promising in the treatment of different neuropsychiatric disorders, such as depression, anxiety, post-traumatic stress disorder (PTSD), addiction, neurodegenerative disorders and autism spectrum disorder (ASD). Given the advances of cannabinoids, especially CBD, in the treatment of epilepsy, would the same expectation regarding the treatment of other neuropsychiatric disorders be possible? The present review highlights some contributions from Brazilian researchers and other studies reported elsewhere on the history, pre-clinical and clinical data underlying the use of cannabinoids for the already widespread treatment of refractory epilepsies and the possibility of use in the treatment of some neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rafael M. Bitencourt
- Laboratory of Behavioral Neuroscience, Graduate Program in Health Sciences, University of Southern Santa Catarina, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Reinaldo N. Takahashi
- Post Graduate Program in Pharmacology, Department of Pharmacology, Federal University of Santa Catarina, Federal University of Santa Catarina (UFSC), Florianópolis, Brazil
| | - Elisaldo A. Carlini
- Centro Brasileiro de Informações Sobre Drogas Psicotrópicas (CEBRID), Department of Preventive Medicine, Federal University of São Paulo, UNIFESP, São Paulo, Brazil
| |
Collapse
|
35
|
Pavón FJ, Polis IY, Stouffer DG, Cravatt BF, Roberto M, Martin-Fardon R, Rodríguez de Fonseca F, Parsons LH, Serrano A. Selective inhibition of monoacylglycerol lipase is associated with passive coping behavior and attenuation of stress-induced dopamine release in the medial prefrontal cortex. Neurobiol Stress 2021; 14:100293. [PMID: 33490317 PMCID: PMC7809503 DOI: 10.1016/j.ynstr.2021.100293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/03/2020] [Accepted: 01/04/2021] [Indexed: 12/18/2022] Open
Abstract
The endocannabinoid system is involved in the regulation of the stress response, but the relative contribution of N-arachidonoylethanolamine (AEA) and 2-arachidonoylglycerol (2-AG) and their mechanisms have to be elucidated. In this study, we compared the effects of the pharmacological inhibition of the two major endocannabinoid-degrading enzymes [fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) for AEA and 2-AG, respectively] on stress-coping [forced swim test (FST) and tail suspension test (TST)] and anxiety-like [elevated-plus maze (EPM) and light-dark test (LDT)] behaviors in wild-type and FAAH knockout mice. In vivo microdialysis estimated the effects of FAAH and MAGL inhibition on dopamine (DA) and serotonin (5-HT) levels in the medial prefrontal cortex (mPFC) during an FST. Mice were treated with PF-3845 (FAAH inhibitor), JZL184 (MAGL inhibitor), JZL195 (dual FAAH/MAGL inhibitor) or vehicle. Our data showed that PF-3845 increased latency to immobility and decreased total immobility time in FST, but no effects were observed in TST compared with vehicle-treated wild-type mice. By contrast, JZL184 decreased latency and increased immobility in TST and FST. JZL195 in wild-type mice and JZL184 in FAAH knockout mice reproduced the same passive coping behaviors as JZL184 in wild-type mice in TST and FST. In the microdialysis experiment, FST was associated with increased DA and 5-HT levels in the mPFC. However, JZL184-treated wild-type mice displayed a significant attenuation of forced swim stress-induced DA release compared with vehicle-treated wild-type mice and PF-3845-treated wild-type mice. Finally, FAAH and/or MAGL inhibitors induced robust and consistent anxiolytic-like effects in EPM and LDT. These results suggested differences between FAAH and MAGL inhibition in stress-coping behaviors. Notably, MAGL inhibition induced a consistent avoidant coping behavior and attenuated the stress-induced mPFC DA response in FST. However, more investigation is needed to elucidate the functional association between DA and 2-AG signaling pathways, and the molecular mechanism in the regulation of passive coping strategies during inescapable stress. FAAH and/or MAGL inhibition induce opposite changes in stress-coping behaviors. MAGL inhibition increases passive stress-coping behaviors in mice. Passive stress-coping behaviors are regulated by 2-AG rather than AEA signaling. MAGL inhibition attenuates mPFC dopamine increase in the forced swim test. FAAH and/or MAGL inhibitors are associated with anxiolytic-like effects.
Collapse
Affiliation(s)
- Francisco Javier Pavón
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA.,Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain.,CIBERCV-Instituto de Salud Carlos III and Unidad de Gestión Clínica del Corazón, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Ilham Y Polis
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - David G Stouffer
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Rémi Martin-Fardon
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Loren H Parsons
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Antonia Serrano
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA.,Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| |
Collapse
|
36
|
Paraíso-Luna J, Aguareles J, Martín R, Ayo-Martín AC, Simón-Sánchez S, García-Rincón D, Costas-Insua C, García-Taboada E, de Salas-Quiroga A, Díaz-Alonso J, Liste I, Sánchez-Prieto J, Cappello S, Guzmán M, Galve-Roperh I. Endocannabinoid signalling in stem cells and cerebral organoids drives differentiation to deep layer projection neurons via CB 1 receptors. Development 2020; 147:226034. [PMID: 33168583 DOI: 10.1242/dev.192161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022]
Abstract
The endocannabinoid (eCB) system, via the cannabinoid CB1 receptor, regulates neurodevelopment by controlling neural progenitor proliferation and neurogenesis. CB1 receptor signalling in vivo drives corticofugal deep layer projection neuron development through the regulation of BCL11B and SATB2 transcription factors. Here, we investigated the role of eCB signalling in mouse pluripotent embryonic stem cell-derived neuronal differentiation. Characterization of the eCB system revealed increased expression of eCB-metabolizing enzymes, eCB ligands and CB1 receptors during neuronal differentiation. CB1 receptor knockdown inhibited neuronal differentiation of deep layer neurons and increased upper layer neuron generation, and this phenotype was rescued by CB1 re-expression. Pharmacological regulation with CB1 receptor agonists or elevation of eCB tone with a monoacylglycerol lipase inhibitor promoted neuronal differentiation of deep layer neurons at the expense of upper layer neurons. Patch-clamp analyses revealed that enhancing cannabinoid signalling facilitated neuronal differentiation and functionality. Noteworthy, incubation with CB1 receptor agonists during human iPSC-derived cerebral organoid formation also promoted the expansion of BCL11B+ neurons. These findings unveil a cell-autonomous role of eCB signalling that, via the CB1 receptor, promotes mouse and human deep layer cortical neuron development.
Collapse
Affiliation(s)
- Juan Paraíso-Luna
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - José Aguareles
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Ricardo Martín
- Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Ane C Ayo-Martín
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.,International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
| | - Samuel Simón-Sánchez
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Daniel García-Rincón
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Carlos Costas-Insua
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Elena García-Taboada
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Adán de Salas-Quiroga
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Javier Díaz-Alonso
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Isabel Liste
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain
| | - José Sánchez-Prieto
- Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | | | - Manuel Guzmán
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Ismael Galve-Roperh
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| |
Collapse
|
37
|
Fonseca R, Madeira N, Simoes C. Resilience to fear: The role of individual factors in amygdala response to stressors. Mol Cell Neurosci 2020; 110:103582. [PMID: 33346000 DOI: 10.1016/j.mcn.2020.103582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/13/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022] Open
Abstract
Resilience to stress is an adaptive process that varies individually. Resilience refers to the adaptation, or the ability to maintain or regain mental health, despite being subject to adverse situation. Resilience is a dynamic concept that reflects a combination of internal individual factors, including age and gender interacting with external factors such as social, cultural and environmental factors. In the last decade, we have witnessed an increase in the prevalence of anxiety disorders, including post-traumatic stress disorder. Given that stress in unavoidable, it is of great interest to understand the neurophysiological mechanisms of resilience, the individual factors that may contribute to susceptibility and promote efficacious approaches to improve resilience. Here, we address this complex question, attempting at defining clear and operational definitions that may allow us to improve our analysis of behavior incorporating individuality. We examine how individual perception of the stressor can alter the outcome of an adverse situation using as an example, the fear-conditioning paradigm and discuss how individual differences in the reward system can contribute to resilience. Given the central role of the endocannabinoid system in regulating fear responses and anxiety, we discuss the evidence that polymorphisms in several molecules of this signaling system contribute to different anxiety phenotypes. The endocannabinoid system is highly interconnected with the serotoninergic and dopaminergic modulatory systems, contributing to individual differences in stress perception and coping mechanisms. We review how the individual variability in these modulatory systems can be used towards a multivariable assessment of stress risk. Incorporating individuality in our research will allow us to define biomarkers of anxiety disorders as well as assess prognosis, towards a personalized clinical approach to mental health.
Collapse
Affiliation(s)
- Rosalina Fonseca
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal.
| | - Natália Madeira
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal
| | - Carla Simoes
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal
| |
Collapse
|
38
|
Abstract
Cannabinoids have been known as the primary component of cannabis for decades, but the characterization of the endocannabinoid system (ECS) in the 1990s opened the doors for cannabis' use in modern medicine. The 2 main receptors of this system, cannabinoid receptors 1 and 2, are found on cells of various tissues, with significant expression in the gastrointestinal (GI) tract. The characterization of the ECS also heralded the understanding of endocannabinoids, naturally occurring compounds synthesized in the human body. Via secondary signaling pathways acting on vagal nerves, nociceptors, and immune cells, cannabinoids have been shown to have both palliative and detrimental effects on the pathophysiology of GI disorders. Although research on the effects of both endogenous and exogenous cannabinoids has been slow due to the complicated legal history of cannabis, discoveries of cannabinoids' treatment potential have been found in various fields of medicine, including the GI world. Medical cannabis has since been offered as a treatment for a myriad of conditions and malignancies, including cancer, human immunodeficiency virus/acquired immunodeficiency syndrome, multiple sclerosis, chronic pain, nausea, posttraumatic stress disorder, amyotrophic lateral sclerosis, cachexia, glaucoma, and epilepsy. This article hopes to create an overview of current research on cannabinoids and the ECS, detail the potential advantages and pitfalls of their use in GI diseases, and explore possible future developments in this field.
Collapse
|
39
|
deRoon-Cassini TA, Stollenwerk TM, Beatka M, Hillard CJ. Meet Your Stress Management Professionals: The Endocannabinoids. Trends Mol Med 2020; 26:953-968. [PMID: 32868170 PMCID: PMC7530069 DOI: 10.1016/j.molmed.2020.07.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/24/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
Abstract
The endocannabinoid signaling system (ECSS) is altered by exposure to stress and mediates and modulates the effects of stress on the brain. Considerable preclinical data support critical roles for the endocannabinoids and their target, the CB1 cannabinoid receptor, in the adaptation of the brain to repeated stress exposure. Chronic stress exposure increases vulnerability to mental illness, so the ECSS has attracted attention as a potential therapeutic target for the prevention and treatment of stress-related psychopathology. We discuss human genetic studies indicating that the ECSS contributes to risk for mental illness in those exposed to severe stress and trauma early in life, and we explore the potential difficulties in pharmacological manipulation of the ECSS.
Collapse
Affiliation(s)
- Terri A deRoon-Cassini
- Neuroscience Research Center, USA; Department of Surgery, Division of Trauma and Acute Care Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Todd M Stollenwerk
- Neuroscience Research Center, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Margaret Beatka
- Neuroscience Research Center, USA; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cecilia J Hillard
- Neuroscience Research Center, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
40
|
Activation of tyrosine phosphatase PTP1B in pyramidal neurons impairs endocannabinoid signaling by tyrosine receptor kinase trkB and causes schizophrenia-like behaviors in mice. Neuropsychopharmacology 2020; 45:1884-1895. [PMID: 32610340 PMCID: PMC7608138 DOI: 10.1038/s41386-020-0755-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022]
Abstract
Schizophrenia is a debilitating disorder affecting young adults displaying symptoms of cognitive impairment, anxiety, and early social isolation prior to episodes of auditory hallucinations. Cannabis use has been tied to schizophrenia-like symptoms, indicating that dysregulated endogenous cannabinoid signaling may be causally linked to schizophrenia. Previously, we reported that glutamatergic neuron-selective ablation of Lmo4, an endogenous inhibitor of the tyrosine phosphatase PTP1B, impairs endocannabinoid (eCB) production from the metabotropic glutamate receptor mGluR5. These Lmo4-deficient mice display anxiety-like behaviors that are alleviated by local shRNA knockdown or pharmacological inhibition of PTP1B that restores mGluR5-dependent eCB production in the amygdala. Here, we report that these Lmo4-deficient mice also display schizophrenia-like behaviors: impaired working memory assessed in the Y maze and defective sensory gating by prepulse inhibition of the acoustic startle response. Modulation of inhibitory inputs onto layer 2/3 pyramidal neurons of the prefrontal cortex relies on eCB signaling from the brain-derived neurotrophic factor receptor trkB, rather than mGluR5, and this mechanism was defective in Lmo4-deficient mice. Genetic ablation of PTP1B in the glutamatergic neurons lacking Lmo4 restored tyrosine phosphorylation of trkB, trkB-mediated eCB signaling, and ameliorated schizophrenia-like behaviors. Pharmacological inhibition of PTP1B with trodusquemine also restored trkB phosphorylation and improved schizophrenia-like behaviors by restoring eCB signaling, since the CB1 receptor antagonist 1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-1-piperidinyl-1H-pyrazole-3-carboxamide blocked this effect. Thus, activation of PTP1B in pyramidal neurons contributes to schizophrenia-like behaviors in Lmo4-deficient mice and genetic or pharmacological intervention targeting PTP1B ameliorates schizophrenia-related deficits.
Collapse
|
41
|
Schuele LL, Glasmacher S, Gertsch J, Roggan MD, Transfeld JL, Bindila L, Lutz B, Kolbe CC, Bilkei-Gorzo A, Zimmer A, Leidmaa E. Diacylglycerol lipase alpha in astrocytes is involved in maternal care and affective behaviors. Glia 2020; 69:377-391. [PMID: 32876968 DOI: 10.1002/glia.23903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 01/05/2023]
Abstract
Genetic deletion of cannabinoid CB1 receptors or diacylglycerol lipase alpha (DAGLa), the main enzyme involved in the synthesis of the endocannabinoid (eCB) 2-arachidonoylglycerol (2-AG), produced profound phenotypes in animal models of depression-related behaviors. Furthermore, clinical studies have shown that antagonists of CB1 can increase the incidence and severity of major depressive episodes. However, the underlying pathomechanisms are largely unknown. In this study, we have focused on the possible involvement of astrocytes. Using the highly sensitive RNAscope technology, we show for the first time that a subpopulation of astrocytes in the adult mouse brain expresses Dagla, albeit at low levels. Targeted lipidomics revealed that astrocytic DAGLa only accounts for a minor percentage of the steady-state brain 2-AG levels and other arachidonic acid derived lipids like prostaglandins. Nevertheless, the deletion of Dagla in adult mouse astrocytes had profound behavioral consequences with significantly increased depressive-like behavioral responses and striking effects on maternal behavior, corresponding with increased levels of serum progesterone and estradiol. Our findings therefore indicate that lipids from the DAGLa metabolic axis in astrocytes play a key regulatory role in affective behaviors.
Collapse
Affiliation(s)
- Lena-Louise Schuele
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sandra Glasmacher
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Marie Denise Roggan
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn, Germany
| | - Janis-Lisa Transfeld
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn, Germany
| | - Laura Bindila
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | | | - Andras Bilkei-Gorzo
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn, Germany
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn, Germany
| | - Este Leidmaa
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
42
|
The endocannabinoid 2-arachidonoylglycerol inhibits endothelial function and repair. Int J Cardiol 2020; 323:243-250. [PMID: 32810540 DOI: 10.1016/j.ijcard.2020.08.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/18/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Endothelial dysfunction promotes atherogenesis, vascular inflammation, and thrombus formation. Reendothelialization after angioplasty is required in order to prevent stent failure. Previous studies have highlighted the role of 2-arachidonoylglycerol (2-AG) in murine experimental atherogenesis and in human coronary artery disease. However, the impact of 2-AG on endothelial repair and leukocyte-endothelial cell adhesion is still unknown. METHODS Endothelial repair was studied in two treatment groups of wildtype mice following electrical injury of the common carotid artery. One group received the monoacylglycerol lipase (MAGL)-inhibitor JZL184, which impairs 2-AG degradation and thus causes elevated 2-AG levels, the other group received DMSO (vehicle). The effect of 2-AG on human coronary artery endothelial cell (HCAEC) viability, leukocyte-endothelial cell adhesion, surface expression of adhesion molecules, and expression of endothelial NO synthase (NOS3) was studied in vitro. RESULTS Elevated 2-AG levels significantly impaired reendothelialization in wildtype mice following electrical injury of the common carotid artery. In vitro, 2-AG significantly reduced viability of HCAEC. Additionally, 2-AG promoted adhesion of THP-1 monocytes to HCAEC following pre-treatment of the HCAEC with 2-AG. Adhesion molecules (E-selectin, ICAM-1 and VCAM-1) remained unchanged in arterial endothelial cells, whereas 2-AG suppressed the expression of NOS3 in HCAEC. CONCLUSION AND TRANSLATIONAL ASPECT Elevated 2-AG levels hamper endothelial repair and HCAEC proliferation, while simultaneously facilitating leukocyte-endothelial cell adhesion. Given that 2-AG is elevated in patients with coronary artery disease and non-ST-segment elevation myocardial infarction, 2-AG might decrease reendothelialization after angioplasty and thus impact the clinical outcomes.
Collapse
|
43
|
Yamasaki T, Ohya T, Mori W, Zhang Y, Wakizaka H, Nengaki N, Fujinaga M, Kikuchi T, Zhang MR. Development of an In Vivo Method to Estimate Effective Drug Doses and Quantify Fatty Acid Amide Hydrolase in Rodent Brain using Positron Emission Tomography Tracer [ 11C]DFMC. J Pharmacol Exp Ther 2020; 373:353-360. [PMID: 32241809 DOI: 10.1124/jpet.119.263772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/27/2020] [Indexed: 11/22/2022] Open
Abstract
Fatty acid amide hydrolase (FAAH) is a key enzyme in the endocannabinoid system. N-(3,4-Dimethylisoxazol-5-yl)piperazine-4-[4-(2-fluoro-4-[11C]methylphenyl)thiazol-2-yl]-1-carboxamide ([11C]DFMC) was developed as an irreversible-type positron emission tomography (PET) tracer for FAAH. Here, we attempted to noninvasively estimate rate constant k3 (rate of transfer to the specifically-bound compartment) as a direct index for FAAH in the rat brain. First, the two-tissue compartment model analysis including three parameters [K1-k3, two-tissue compartment model for the irreversible-type radiotracer (2TCMi)] in PET study with [11C]DFMC was conducted, which provided 0.21 ± 0.04 ml·cm-3·min-1 of the net uptake value (Ki), an indirect index for FAAH, in the FAAH-richest region (the cingulate cortex). Subsequently, to noninvasively estimate Ki value, the reference model analysis (Patlak graphical analysis reference model) was tried using a time-activity curve of the spinal cord. In that result, the noninvasive Ki value (KREF) was concisely estimated with high correlation (r > 0.95) to Ki values based on 2TCMi. Using estimated KREF value, we tried to obtain calculated-k3 based on previously defined equations. The calculated k3 was successfully estimated with high correlation (r = 0.95) to direct k3 in 2TCMi. Finally, the dose relationship study using calculated k3 demonstrated that in vivo ED50 value of [3-(3-carbamoylphenyl)phenyl] N-cyclohexylcarbamate, a major inhibitor of FAAH, was 66.4 µg/kg in rat brain. In conclusion, we proposed the calculated k3 as an alternative index corresponding to regional FAAH concentrations and suggested that PET with [11C]DFMC enables occupancy study for new pharmaceuticals targeting FAAH. SIGNIFICANCE STATEMENT: In the present study, we proposed calculated k3 as an alternative index corresponding with fatty acid amide hydrolase concentration. By using calculated k3, in vivo ED50 of [3-(3-carbamoylphenyl)phenyl] N-cyclohexylcarbamate was successfully estimated to be 66.4 µg/kg for rats. Thus, we demonstrated the pharmacological utility of positron emission tomography with N-(3,4-dimethylisoxazol-5-yl)piperazine-4-[4-(2-fluoro-4-[11C]methylphenyl)thiazol-2-yl]-1-carboxamide.
Collapse
Affiliation(s)
- Tomoteru Yamasaki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (T.Y., T.O., W.M., Y.Z., H.W., N.N., M.F., T.K., M.-R.Z.) and SHI Accelerator Service Co. Ltd, Tokyo, Japan (N.N.)
| | - Tomoyuki Ohya
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (T.Y., T.O., W.M., Y.Z., H.W., N.N., M.F., T.K., M.-R.Z.) and SHI Accelerator Service Co. Ltd, Tokyo, Japan (N.N.)
| | - Wakana Mori
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (T.Y., T.O., W.M., Y.Z., H.W., N.N., M.F., T.K., M.-R.Z.) and SHI Accelerator Service Co. Ltd, Tokyo, Japan (N.N.)
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (T.Y., T.O., W.M., Y.Z., H.W., N.N., M.F., T.K., M.-R.Z.) and SHI Accelerator Service Co. Ltd, Tokyo, Japan (N.N.)
| | - Hidekatsu Wakizaka
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (T.Y., T.O., W.M., Y.Z., H.W., N.N., M.F., T.K., M.-R.Z.) and SHI Accelerator Service Co. Ltd, Tokyo, Japan (N.N.)
| | - Nobuki Nengaki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (T.Y., T.O., W.M., Y.Z., H.W., N.N., M.F., T.K., M.-R.Z.) and SHI Accelerator Service Co. Ltd, Tokyo, Japan (N.N.)
| | - Masayuki Fujinaga
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (T.Y., T.O., W.M., Y.Z., H.W., N.N., M.F., T.K., M.-R.Z.) and SHI Accelerator Service Co. Ltd, Tokyo, Japan (N.N.)
| | - Tatsuya Kikuchi
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (T.Y., T.O., W.M., Y.Z., H.W., N.N., M.F., T.K., M.-R.Z.) and SHI Accelerator Service Co. Ltd, Tokyo, Japan (N.N.)
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (T.Y., T.O., W.M., Y.Z., H.W., N.N., M.F., T.K., M.-R.Z.) and SHI Accelerator Service Co. Ltd, Tokyo, Japan (N.N.)
| |
Collapse
|
44
|
Cannabinoid agonists possibly mediate interaction between cholinergic and cannabinoid systems in regulating intestinal inflammation. Med Hypotheses 2020; 139:109613. [DOI: 10.1016/j.mehy.2020.109613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/31/2020] [Accepted: 02/04/2020] [Indexed: 02/07/2023]
|
45
|
Protective effects of elevated anandamide on stress and fear-related behaviors: translational evidence from humans and mice. Mol Psychiatry 2020; 25:993-1005. [PMID: 30120421 DOI: 10.1038/s41380-018-0215-1] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 01/08/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a common, debilitating condition with limited treatment options. Extinction of fear memories through prolonged exposure therapy, the primary evidence-based behavioral treatment for PTSD, has only partial efficacy. In mice, pharmacological inhibition of fatty acid amide hydrolase (FAAH) produces elevated levels of anandamide (AEA) and promotes fear extinction, suggesting that FAAH inhibitors may aid fear extinction-based treatments. A human FAAH 385C->A substitution encodes an FAAH enzyme with reduced catabolic efficacy. Individuals homozygous for the FAAH 385A allele may therefore offer a genetic model to evaluate the impact of elevations in AEA signaling in humans, helping to inform whether FAAH inhibitors have the potential to facilitate fear extinction therapy for PTSD. To overcome the challenge posed by low frequency of the AA genotype (appr. 5%), we prospectively genotyped 423 individuals to examine the balanced groups of CC, AC, and AA individuals (n = 25/group). Consistent with its loss-of-function nature, the A allele was dose dependently associated with elevated basal AEA levels, facilitated fear extinction, and enhanced the extinction recall. Moreover, the A-allele homozygotes were protected against stress-induced decreases in AEA and negative emotional consequences of stress. In a humanized mouse model, AA homozygous mice were similarly protected against stress-induced decreases in AEA, both in the periphery, and also in the amygdala and prefrontal cortex, brain structures critically involved in fear extinction and regulation of stress responses. Collectively, these data suggest that AEA signaling can temper aspects of the stress response and that FAAH inhibition may aid the treatment for stress-related psychiatric disorders, such as PTSD.
Collapse
|
46
|
Endocannabinoid 2-arachidonoylglycerol is elevated in the coronary circulation during acute coronary syndrome. PLoS One 2019; 14:e0227142. [PMID: 31887202 PMCID: PMC6936850 DOI: 10.1371/journal.pone.0227142] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/12/2019] [Indexed: 11/19/2022] Open
Abstract
Objectives The endocannabinoid system modulates coronary circulatory function and atherogenesis. The two major endocannabinoids (eCB), 2-arachidonoylglycerol (2-AG) and N-arachidonoylethanolamide (AEA), are increased in venous blood from patients with coronary artery disease (CAD). However, given their short half-life and their autocrine/paracrine mechanism of action, eCB levels in venous blood samples might not reflect arterial or coronary eCB concentrations. The aim of this cross-sectional study was to identify the local concentration profile of eCB and to detect whether and how this concentration profile changes in CAD and NSTEMI versus patients without CAD. Methods and results 83 patients undergoing coronary angiography were included in this study. Patients were divided into three groups based on their definite diagnosis of a) no CAD, b) stable CAD, or c) non-ST-segment elevation myocardial infarction (NSTEMI). Blood was drawn from the arterial sheath and the aorta in all patients and additionally distal to the culprit coronary lesion in CAD- and NSTEMI patients. 2-AG levels varied significantly between patient groups and between the sites of blood extraction. The lowest levels were detected in patients without CAD; the highest 2-AG concentrations were detected in NSTEMI patients and in the coronary arteries. Peripheral 2-AG levels were significantly higher in NSTEMI patients (107.4 ± 28.4 pmol/ml) than in CAD- (17.4 ± 5.4 pmol/ml; p < 0.001), or no-CAD patients (23.9 ± 7.1 pmol/ml; p < 0.001). Moreover, coronary 2-AG levels were significantly higher in NSTEMI patients than in CAD patients (369.3 ± 57.2 pmol/ml vs. 240.1 ± 25.3 pmol/ml; p = 0.024). Conclusions 2-AG showed significant variability in arterial blood samples drawn from distinct locations. Possibly, lesional macrophages synthesise 2-AG locally, which thereby contributes to endothelial dysfunction and local inflammation.
Collapse
|
47
|
Stress-induced modulation of endocannabinoid signaling leads to delayed strengthening of synaptic connectivity in the amygdala. Proc Natl Acad Sci U S A 2019; 117:650-655. [PMID: 31843894 DOI: 10.1073/pnas.1910322116] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Even a brief exposure to severe stress strengthens synaptic connectivity days later in the amygdala, a brain area implicated in the affective symptoms of stress-related psychiatric disorders. However, little is known about the synaptic signaling mechanisms during stress that eventually culminate in its delayed impact on the amygdala. Hence, we investigated early stress-induced changes in amygdalar synaptic signaling in order to prevent its delayed effects. Whole-cell recordings in basolateral amygdala (BLA) slices from rats revealed higher frequency of miniature excitatory postsynaptic currents (mEPSCs) immediately after 2-h immobilization stress. This was replicated by inhibition of cannabinoid receptors (CB1R), suggesting a role for endocannabinoid (eCB) signaling. Stress also reduced N-arachidonoylethanolamine (AEA), an endogenous ligand of CB1R. Since stress-induced activation of fatty acid amide hydrolase (FAAH) reduces AEA, we confirmed that oral administration of an FAAH inhibitor during stress prevents the increase in synaptic excitation in the BLA soon after stress. Although stress also caused an immediate reduction in synaptic inhibition, this was not prevented by FAAH inhibition. Strikingly, FAAH inhibition during the traumatic stressor was also effective 10 d later on the delayed manifestation of synaptic strengthening in BLA neurons, preventing both enhanced mEPSC frequency and increased dendritic spine-density. Thus, oral administration of an FAAH inhibitor during a brief stress prevents the early synaptic changes that eventually build up to hyperexcitability in the amygdala. This framework is of therapeutic relevance because of growing interest in targeting eCB signaling to prevent the gradual development of emotional symptoms and underlying amygdalar dysfunction triggered by traumatic stress.
Collapse
|
48
|
Monoacylglycerol Lipase Inactivation by Using URB602 Mitigates Myocardial Damage in a Rat Model of Cardiac Arrest. Crit Care Med 2019; 47:e144-e151. [PMID: 30431495 DOI: 10.1097/ccm.0000000000003552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Monoacylglycerol lipase participates in organ protection by regulating the hydrolysis of the endocannabinoid 2-arachidonoylglycerol. This study investigated whether blocking monoacylglycerol lipase protects against postresuscitation myocardial injury and improves survival in a rat model of cardiac arrest and cardiopulmonary resuscitation. DESIGN Prospective randomized laboratory study. SETTING University research laboratory. SUBJECTS Male Sprague-Dawley rat (n = 96). INTERVENTIONS Rats underwent 8-minute asphyxia-based cardiac arrest and resuscitation. Surviving rats were randomly divided into cardiopulmonary resuscitation + URB602 group, cardiopulmonary resuscitation group, and sham group. One minute after successful resuscitation, rats in the cardiopulmonary resuscitation + URB602 group received a single dose of URB602 (5 mg/kg), a small-molecule monoacylglycerol lipase inhibitor, whereas rats in the cardiopulmonary resuscitation group received an equivalent volume of vehicle solution. The sham rats underwent all of the procedures performed on rats in the cardiopulmonary resuscitation and cardiopulmonary resuscitation + URB602 groups minus cardiac arrest and asphyxia. MEASUREMENTS AND MAIN RESULTS Survival was recorded 168 hours after the return of spontaneous circulation (n = 22 in each group). Compared with vehicle treatment (31.8%), URB602 treatment markedly improved survival (63.6%) 168 hours after cardiopulmonary resuscitation. Next, we used additional surviving rats to evaluate myocardial and mitochondrial injury 6 hours after return of spontaneous circulation, and we found that URB602 significantly reduced myocardial injury and prevented myocardial mitochondrial damage. In addition, URB602 attenuated the dysregulation of endocannabinoid and eicosanoid metabolism 6 hours after return of spontaneous circulation and prevented the acceleration of mitochondrial permeability transition 15 minutes after return of spontaneous circulation. CONCLUSIONS Monoacylglycerol lipase blockade may reduce myocardial and mitochondrial injury and significantly improve the resuscitation effect after cardiac arrest and cardiopulmonary resuscitation.
Collapse
|
49
|
Fitzgibbon M, Kerr DM, Henry RJ, Finn DP, Roche M. Endocannabinoid modulation of inflammatory hyperalgesia in the IFN-α mouse model of depression. Brain Behav Immun 2019; 82:372-381. [PMID: 31505257 DOI: 10.1016/j.bbi.2019.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022] Open
Abstract
Depression is a well-recognised effect of long-term treatment with interferon-alpha (IFN-α), a widely used treatment for chronic viral hepatitis and malignancy. In addition to the emotional disturbances, high incidences of painful symptoms such as headache and joint pain have also been reported following IFN-α treatment. The endocannabinoid system plays an important role in emotional and nociceptive processing, however it is unknown whether repeated IFN-α administration induces alterations in this system. The present study investigated nociceptive responding in the IFN-α-induced mouse model of depression and associated changes in the endocannabinoid system. Furthermore, the effects of modulating peripheral endocannabinoid tone on inflammatory pain-related behaviour in the IFN-α model was examined. Repeated IFN-α administration (8000 IU/g/day) to male C57/Bl6 mice increased immobility in the forced swim test and reduced sucrose preference, without altering body weight gain or locomotor activity, confirming development of the depressive-like phenotype. There was no effect of repeated IFN-α administration on latency to respond in the hot plate test on day 4 or 7 of treatment, however, formalin-evoked nociceptive behaviour was significantly increased in IFN-α treated mice following 8 days of IFN-α administration. 2-Arachidonoyl glycerol (2-AG) levels in the periaqueductal grey (PAG) and rostroventromedial medulla (RVM), and anandamide (AEA) levels in the RVM, were significantly increased in IFN-α-, but not saline-, treated mice following formalin administration. There was no change in endocannabinoid levels in the prefrontal cortex, spinal cord or paw tissue between saline- or IFNα-treated mice in the presence or absence of formalin. Furthermore, repeated IFN-α and/or formalin administration did not alter mRNA expression of genes encoding the endocannabinoid catabolic enzymes (fatty acid amide hydrolase or monoacylglycerol lipase) or endocannabinoid receptor targets (CB1, CB2 or PPARs) in the brain, spinal cord or paw tissue. Intra plantar administration of PF3845 (1 μg/10 μl) or MJN110 (1 μg/10 μl), inhibitors of AEA and 2-AG catabolism respectively, attenuated formalin-evoked hyperalgesia in IFN-α, but not saline-, treated mice. In summary, increasing peripheral endocannabinoid tone attenuates inflammatory hyperalgesia induced following repeated IFN-α administration. These data provide support for the endocannabinoid system in mediating and modulating heightened pain responding associated with IFNα-induced depression.
Collapse
Affiliation(s)
- Marie Fitzgibbon
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland
| | - Daniel M Kerr
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland; Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, Ireland
| | - Rebecca J Henry
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, Ireland; NCBES Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Michelle Roche
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland; NCBES Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland.
| |
Collapse
|
50
|
Rat ultrasonic vocalizations as a measure of the emotional component of chronic pain. Neuroreport 2019; 30:863-866. [DOI: 10.1097/wnr.0000000000001282] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|