1
|
Kroll SL, Meier P, Mayo LM, Gertsch J, Quednow BB. Endocannabinoids and related lipids linked to social exclusion in individuals with chronic non-medical prescription opioid use. Neuropsychopharmacology 2024; 49:1630-1639. [PMID: 38773316 PMCID: PMC11319498 DOI: 10.1038/s41386-024-01881-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/10/2024] [Accepted: 04/29/2024] [Indexed: 05/23/2024]
Abstract
Opioid-related overdose deaths are still on the rise in North America, emphasizing the need to better understand the underlying neurobiological mechanisms regarding the development of opioid use disorder (OUD). Recent evidence from preclinical and clinical studies indicate that the endocannabinoid system (ECS) may play a crucial role in stress and reward, both involved in the development and maintenance of substance use disorders. Animal models demonstrate a specific crosstalk between the ECS and the endogenous opioid system. However, translational studies in humans are scarce. Here, we investigated basal plasma levels of the endocannabinoids anandamide (AEA) and 2-arachidonoyglycerol (2-AG), and eight endocannabinoid-related lipids, including oleoylethanolamide (OEA) and palmitoylethanolamide (PEA), as well as whole blood fatty acid amide hydrolase (FAAH) activity in chronic non-medical prescription opioid users (NMPOU; n = 21) compared to opioid-naïve healthy controls (n = 29) considering age, sex, and cannabis use as potential confounders. Additionally, the association of endocannabinoids and related lipids with the participants' response to experimentally induced social exclusion was examined. We found significantly elevated basal AEA, OEA, and PEA levels in NMPOU compared to controls, but no differences in FAAH activity, 2-AG, or other endocannabinoid-related lipids. Within NMPOU, higher AEA levels were associated with lower perception of social exclusion. Robust positive correlations within N-acylethanolamines (i.e., AEA, OEA, and PEA) indicate strong metabolic associations. Together with our recent findings of elevated basal 2-AG levels in dependent cocaine users, present results indicate substance-specific alterations of the ECS that may have implications in the search for novel therapeutic interventions for these populations.
Collapse
Affiliation(s)
- Sara L Kroll
- Social and Affective Neuropsychopharmacology, Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland.
- Experimental and Clinical Pharmacopsychology, Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology, Zurich, Switzerland.
| | - Philip Meier
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Leah M Mayo
- Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, and Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Boris B Quednow
- Experimental and Clinical Pharmacopsychology, Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology, Zurich, Switzerland
| |
Collapse
|
2
|
Wang X, Liu Y, Zhou Y, Li M, Mo T, Xu X, Chen Z, Zhang Y, Yang L. mTORC2 knockdown mediates lipid metabolism to alleviate hyperlipidemic pancreatitis through PPARα. J Biochem Mol Toxicol 2024; 38:e23802. [PMID: 39132808 DOI: 10.1002/jbt.23802] [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: 03/19/2024] [Revised: 07/04/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
Hyperlipidemic pancreatitis (HP) is an inflammatory injury of the pancreas triggered by elevated serum triglyceride (TG) levels. The mechanistic target of rapamycin (mTOR) signaling pathway plays a crucial role in regulating lipid homeostasis and inflammation. This study aimed to investigate whether the activity of mTOR complex 2 (mTORC2) affects the progression of HP and its underlying mechanisms. In vivo, a high-fat diet and retrograde administration of sodium taurocholate were employed to establish the HP models in rats, with pancreatic tissue pathology evaluated. The expression of Rictor and peroxisome proliferator-activator receptor (PPAR) was examined. The serum levels of TG, fatty acid metabolites, inflammatory and lipid metabolism-related factors were determined. In vitro, pancreatic acinar cells (PACs) were exposed to palmitic acid and cholecystokinin-8. PAC apoptosis, pyroptosis, and ferroptosis were assessed. In the HP models, rats and PACs exhibited upregulated Rictor and downregulated PPARα, and Rictor knockdown promoted PPARα expression. In vivo, Rictor knockdown decreased the serum levels of TG, α-amylase, total cholesterol, low-density lipoprotein cholesterol, lactate dehydrogenase, and inflammatory factors, while increasing high-density lipoprotein cholesterol levels. Rictor knockdown increased ACOX1 and CPT1α and decreased SREBP-1, CD36, SCD1, ACLY, and ACACA. Rictor knockdown reduced damage to pancreatic tissue structure. In vitro, Rictor knockdown inhibited PAC apoptosis, pyroptosis, and ferroptosis. Treatment with the PPARα antagonist GW6471 abolished the beneficial effects of Rictor knockdown. Rictor/mTORC2 deficiency reduces serum TG levels, maintains lipid homeostasis, and suppresses inflammation by inhibiting PPARα expression. Weakening mTORC2 activity holds promise as a novel therapeutic strategy for HP.
Collapse
Affiliation(s)
- Xiangyang Wang
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yilei Liu
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yaxiong Zhou
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Min Li
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Tingting Mo
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Xiaoping Xu
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Zhiyuan Chen
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yu Zhang
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Li Yang
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| |
Collapse
|
3
|
Fotio Y, Mabou Tagne A, Squire E, Lee HL, Phillips CM, Chang K, Ahmed F, Greenberg AS, Villalta SA, Scarfone VM, Spadoni G, Mor M, Piomelli D. NAAA-regulated lipid signaling in monocytes controls the induction of hyperalgesic priming in mice. Nat Commun 2024; 15:1705. [PMID: 38402219 PMCID: PMC10894261 DOI: 10.1038/s41467-024-46139-5] [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: 08/14/2023] [Accepted: 02/15/2024] [Indexed: 02/26/2024] Open
Abstract
Circulating monocytes participate in pain chronification but the molecular events that cause their deployment are unclear. Using a mouse model of hyperalgesic priming (HP), we show that monocytes enable progression to pain chronicity through a mechanism that requires transient activation of the hydrolase, N-acylethanolamine acid amidase (NAAA), and the consequent suppression of NAAA-regulated lipid signaling at peroxisome proliferator-activated receptor-α (PPAR-α). Inhibiting NAAA in the 72 hours following administration of a priming stimulus prevented HP. This effect was phenocopied by NAAA deletion and depended on PPAR-α recruitment. Mice lacking NAAA in CD11b+ cells - monocytes, macrophages, and neutrophils - were resistant to HP induction. Conversely, mice overexpressing NAAA or lacking PPAR-α in the same cells were constitutively primed. Depletion of monocytes, but not resident macrophages, generated mice that were refractory to HP. The results identify NAAA-regulated signaling in monocytes as a control node in the induction of HP and, potentially, the transition to pain chronicity.
Collapse
Affiliation(s)
- Yannick Fotio
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Alex Mabou Tagne
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Erica Squire
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Connor M Phillips
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Kayla Chang
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Faizy Ahmed
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | | | - S Armando Villalta
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Vanessa M Scarfone
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, USA
| | - Gilberto Spadoni
- Dipartimento di Scienze Biomolecolari, Università di Urbino "Carlo Bo,", Urbino, Italy
| | - Marco Mor
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA.
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
4
|
Ahmed F, Torrens A, Mahler SV, Ferlenghi F, Huestis MA, Piomelli D. A Sensitive Ultrahigh-Performance Liquid Chromatography/Tandem Mass Spectrometry Method for the Simultaneous Analysis of Phytocannabinoids and Endocannabinoids in Plasma and Brain. Cannabis Cannabinoid Res 2024; 9:371-385. [PMID: 36367975 PMCID: PMC10874825 DOI: 10.1089/can.2022.0216] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Introduction: Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) are major chemical constituents of cannabis, which may interact either directly or indirectly with the endocannabinoid and endocannabinoid-like ("paracannabinoid") systems, two lipid-based signaling complexes that play important roles in physiology. Legislative changes emphasize the need to understand how THC and CBD might impact endocannabinoid and paracannabinoid signaling, and to develop analytical approaches to study such impact. In this study, we describe a sensitive and accurate method for the simultaneous quantification of THC, its main oxidative metabolites [11-hydroxy-Δ9-THC (11-OH-THC) and 11-nor-9-carboxy-Δ9-THC (11-COOH-THC)], CBD, and a representative set of endocannabinoid [anandamide and 2-arachidonoyl-sn-glycerol (2-AG)] and paracannabinoid [palmitoylethanolamide (PEA) and oleoylethanolamide (OEA)] compounds. Analyte separation relies on the temperature-dependent shape selectivity properties of polymerically bonded C18 stationary phases. Materials and Methods: Analytes are extracted from tissues using acetonitrile precipitation followed by phospholipid removal. The ultrahigh-performance liquid chromatography/tandem mass spectrometry protocol utilizes a commercially available C18 polymeric-bonded phase column and a simple gradient elution system. Results: Ten-point calibration curves show excellent linearity (R2>0.99) over a wide range of analyte concentrations (0.02-500 ng/mL). Lowest limits of quantification are 0.05 ng/mL for anandamide, 0.1 ng/mL for 11-OH-THC and OEA, 0.2 ng/mL for THC and CBD, 0.5 ng/mL for 11-COOH-THC, 1.0 ng/mL for 2-AG, and 2.0 ng/mL for PEA. The lowest limits of detection are 0.02 ng/mL for anandamide, 0.05 ng/mL for 11-OH-THC and OEA, 0.1 ng/mL for THC and CBD, 0.2 ng/mL for 11-COOH-THC, 0.5 ng/mL for 2-AG, and 1.0 ng/mL for PEA. Conclusions: An application of the method is presented, which showed that phytocannabinoid administration elevates endocannabinoid levels in plasma and brain of adolescent male and female mice.
Collapse
Affiliation(s)
- Faizy Ahmed
- Department of Anatomy and Neurobiology, University of California, Irvine, California, USA
| | - Alexa Torrens
- Department of Anatomy and Neurobiology, University of California, Irvine, California, USA
| | - Stephen V. Mahler
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
| | - Francesca Ferlenghi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parma, Italy
| | - Marilyn A. Huestis
- Institute of Emerging Health Professions, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, California, USA
- Department of Biological Chemistry, University of California, Irvine, California, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, California, USA
| |
Collapse
|
5
|
Lee MT, Mackie K, Chiou LC. Alternative pain management via endocannabinoids in the time of the opioid epidemic: Peripheral neuromodulation and pharmacological interventions. Br J Pharmacol 2023; 180:894-909. [PMID: 34877650 PMCID: PMC9170838 DOI: 10.1111/bph.15771] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 11/23/2021] [Accepted: 11/27/2021] [Indexed: 01/18/2023] Open
Abstract
The use of opioids in pain management is hampered by the emergence of analgesic tolerance, which leads to increased dosing and side effects, both of which have contributed to the opioid epidemic. One promising potential approach to limit opioid analgesic tolerance is activating the endocannabinoid system in the CNS, via activation of CB1 receptors in the descending pain inhibitory pathway. In this review, we first discuss preclinical and clinical evidence revealing the potential of pharmacological activation of CB1 receptors in modulating opioid tolerance, including activation by phytocannabinoids, synthetic CB1 receptor agonists, endocannabinoid degradation enzyme inhibitors, and recently discovered positive allosteric modulators of CB1 receptors. On the other hand, as non-pharmacological pain relief is advocated by the US-NIH to combat the opioid epidemic, we also discuss contributions of peripheral neuromodulation, involving the electrostimulation of peripheral nerves, in addressing chronic pain and opioid tolerance. The involvement of supraspinal endocannabinoid systems in peripheral neuromodulation-induced analgesia is also discussed. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Grants
- MOST 108-2321-B-002-005 Ministry of Science and Technology, Taiwan
- MOST 107-2811-B-002-008 Ministry of Science and Technology, Taiwan
- R01 DA041229 NIDA NIH HHS
- MOST 107-2321-B-002-010 Ministry of Science and Technology, Taiwan
- R01 DA047858 NIDA NIH HHS
- 107M4022-3 Ministry of Education, Taiwan
- MOST 106-2321-B-002-019 Ministry of Science and Technology, Taiwan
- NHRI-EX111-11114NI National Health Research Institutes, Taiwan
- FRGS/1/2021/WAB13/UCSI/02/1 Ministry of Higher Education, Malaysia
- R21 DA042584 NIDA NIH HHS
- REIG-FPS-2020/065 UCSI University Research Excellence and Innovation Grant, Malaysia
- NHRI-EX109-10733NI National Health Research Institutes, Taiwan
- MOST 104-2745-B-002-004 Ministry of Science and Technology, Taiwan
- MOST 109-2320-B-002-042-MY3 Ministry of Science and Technology, Taiwan
- MOST 107-2811-B-002 -008 Ministry of Science and Technology, Taiwan
- MOST 108-2320-B-002-029-MY3 Ministry of Science and Technology, Taiwan
Collapse
Affiliation(s)
- Ming Tatt Lee
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Ken Mackie
- Gill Center for Biomolecular Research, Indiana University, Bloomington, Indiana 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405, USA
| | - Lih-Chu Chiou
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
6
|
Palese F, Pontis S, Realini N, Torrens A, Ahmed F, Assogna F, Pellicano C, Bossù P, Spalletta G, Green K, Piomelli D. Targeting NAAA counters dopamine neuron loss and symptom progression in mouse models of parkinsonism. Pharmacol Res 2022; 182:106338. [PMID: 35781057 PMCID: PMC9733952 DOI: 10.1016/j.phrs.2022.106338] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/19/2022] [Accepted: 06/29/2022] [Indexed: 12/13/2022]
Abstract
The lysosomal cysteine hydrolase N-acylethanolamine acid amidase (NAAA) deactivates palmitoylethanolamide (PEA), a lipid-derived PPAR-α agonist that is critically involved in the control of pain and inflammation. In this study, we asked whether NAAA-regulated PEA signaling might contribute to dopamine neuron degeneration and parkinsonism induced by the mitochondrial neurotoxins, 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). In vitro experiments showed that 6-OHDA and MPTP enhanced NAAA expression and lowered PEA content in human SH-SY5Y cells. A similar effect was observed in mouse midbrain dopamine neurons following intra-striatal 6-OHDA injection. Importantly, deletion of the Naaa gene or pharmacological inhibition of NAAA activity substantially attenuated both dopamine neuron death and parkinsonian symptoms in mice treated with 6-OHDA or MPTP. Moreover, NAAA expression was elevated in postmortem brain cortex and premortem blood-derived exosomes from persons with Parkinson's disease compared to age-matched controls. The results identify NAAA-regulated PEA signaling as a molecular control point for dopaminergic neuron survival and a potential target for neuroprotective intervention.
Collapse
Affiliation(s)
- Francesca Palese
- Department of Anatomy and Neurobiology University of California Irvine, 92697-1275 CA, USA
| | - Silvia Pontis
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Natalia Realini
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Alexa Torrens
- Department of Anatomy and Neurobiology University of California Irvine, 92697-1275 CA, USA
| | - Faizy Ahmed
- Department of Anatomy and Neurobiology University of California Irvine, 92697-1275 CA, USA
| | - Francesca Assogna
- Laboratorio di Neuropsichiatria, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Clelia Pellicano
- Laboratorio di Neuropsichiatria, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Paola Bossù
- Laboratorio di Neuropsichiatria, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Gianfranco Spalletta
- Laboratorio di Neuropsichiatria, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Kim Green
- Department of Neurobiology and Behavior, University of California Irvine, 92697-1275 CA, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology University of California Irvine, 92697-1275 CA, USA,Department of Pharmaceutical Sciences, University of California Irvine, 92697-1275 CA, USA,Department of Biological Chemistry, University of California Irvine, 92697-1275 CA, USA
| |
Collapse
|
7
|
Abstract
This paper is the forty-third consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2020 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY, 11367, United States.
| |
Collapse
|
8
|
Sihag J, Di Marzo V. (Wh)olistic (E)ndocannabinoidome-Microbiome-Axis Modulation through (N)utrition (WHEN) to Curb Obesity and Related Disorders. Lipids Health Dis 2022; 21:9. [PMID: 35027074 PMCID: PMC8759188 DOI: 10.1186/s12944-021-01609-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/05/2021] [Indexed: 02/06/2023] Open
Abstract
The discovery of the endocannabinoidome (eCBome) is evolving gradually with yet to be elucidated functional lipid mediators and receptors. The diet modulates these bioactive lipids and the gut microbiome, both working in an entwined alliance. Mounting evidence suggests that, in different ways and with a certain specialisation, lipid signalling mediators such as N-acylethanolamines (NAEs), 2-monoacylglycerols (2-MAGs), and N-acyl-amino acids (NAAs), along with endocannabinoids (eCBs), can modulate physiological mechanisms underpinning appetite, food intake, macronutrient metabolism, pain sensation, blood pressure, mood, cognition, and immunity. This knowledge has been primarily utilised in pharmacology and medicine to develop many drugs targeting the fine and specific molecular pathways orchestrating eCB and eCBome activity. Conversely, the contribution of dietary NAEs, 2-MAGs and eCBs to the biological functions of these molecules has been little studied. In this review, we discuss the importance of (Wh) olistic (E)ndocannabinoidome-Microbiome-Axis Modulation through (N) utrition (WHEN), in the management of obesity and related disorders.
Collapse
Affiliation(s)
- Jyoti Sihag
- Faculty of Medicine, University of Laval, Quebec, Canada.
- Faculty of Agriculture and Food Sciences, University of Laval, Quebec, Canada.
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), University of Laval, Quebec, Canada.
- University Institute of Cardiology and Pneumology, Quebec, Canada.
- Institute of Nutrition and Functional Foods (INAF) and Centre Nutrition, Santé et Société (NUTRISS), University of Laval, Quebec, Canada.
- Department of Foods and Nutrition, Chaudhary Charan Singh Haryana Agricultural University, Hisar, India.
| | - Vincenzo Di Marzo
- Faculty of Medicine, University of Laval, Quebec, Canada.
- Faculty of Agriculture and Food Sciences, University of Laval, Quebec, Canada.
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), University of Laval, Quebec, Canada.
- University Institute of Cardiology and Pneumology, Quebec, Canada.
- Institute of Nutrition and Functional Foods (INAF) and Centre Nutrition, Santé et Société (NUTRISS), University of Laval, Quebec, Canada.
- Institute of Biomolecular Chemistry of the National Research Council (ICB-CNR), Naples, Italy.
- Endocannabinoid Research Group, Naples, Italy.
- Joint International Research Unit between the Italian National Research Council (CNR) and University of Laval, for Chemical and Biomolecular Research on the Microbiome and its impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Quebec, Canada.
| |
Collapse
|
9
|
Kong Q, Tian S, Ma C, Wang G, Zhang M. Cannabinoid Receptor Type 2 Agonist Reduces Morphine Tolerance via Mitogen Activated Protein Kinase Phosphatase Induction and Mitogen Activated Protein Kinase Dephosphorylation. Neuroscience 2022; 480:56-64. [PMID: 34774714 DOI: 10.1016/j.neuroscience.2021.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022]
Abstract
Morphine is an opioid drug often used in treating moderate to severe pain. However, morphine tolerance in patients limits its used in clinical settings. Our previous study showed that a cannabinoid type 2 (CB2) receptor agonist attenuated morphine tolerance. However, the exact mechanism by which CB2 agonists reduce morphine tolerance remains unclear. In this study, we investigated the effect of mitogen activated protein kinase (MAPK) and mitogen activated protein kinase phosphatases 1 and 3 (MKP-1 and MKP-3) on the regulation of morphine tolerance by CB2 receptor agonist. Chronic morphine treatments for 7 days reduced the protein expression of MKP-1 and MKP-3 in the spinal cord and increased the phosphorylation of p38, ERK1/2 and the level of proinflammatory mediator, such as IL-1β, IL-6 and TNF-α. Coadministration of CB2 receptor agonist AM1241 alleviated the inhibition of MKP-1 and MKP-3 by chronic morphine administration and reduced the expression of phosphorylated MAPK and proinflammatory factors. The effect of the CB2 receptor agonist on morphine-induced downregulation of MKP-1 and MKP-3 was reversed by the MKP-1 and MKP-3 antagonist triptolide. Our findings suggested that CB2 receptor agonist may induce the expression of MKP-1 and MKP-3 to promote MAPK dephosphorylation and reduce the production of downstream cytokine, thereby reducing morphine tolerance. This finding suggested that MKPs may serve as a new target for alleviating morphine tolerance.
Collapse
Affiliation(s)
- Qingling Kong
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Songyu Tian
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Chao Ma
- Department of Anesthesiology, The Fourth Hospital of Harbin Medical University, Harbin, China.
| | - Guonian Wang
- Department of Anesthesiology, The Fourth Hospital of Harbin Medical University, Harbin, China.
| | - Mingyue Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
10
|
Congiu M, Micheli L, Santoni M, Sagheddu C, Muntoni AL, Makriyannis A, Malamas MS, Ghelardini C, Di Cesare Mannelli L, Pistis M. N-Acylethanolamine Acid Amidase Inhibition Potentiates Morphine Analgesia and Delays the Development of Tolerance. Neurotherapeutics 2021; 18:2722-2736. [PMID: 34553321 PMCID: PMC8804012 DOI: 10.1007/s13311-021-01116-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 11/26/2022] Open
Abstract
Opioids are essential drugs for pain management, although long-term use is accompanied by tolerance, necessitating dose escalation, and dependence. Pharmacological treatments that enhance opioid analgesic effects and/or attenuate the development of tolerance (with a desirable opioid-sparing effect in treating pain) are actively sought. Among them, N-palmitoylethanolamide (PEA), an endogenous lipid neuromodulator with anti-inflammatory and neuroprotective properties, was shown to exert anti-hyperalgesic effects and to delay the emergence of morphine tolerance. A selective augmentation in endogenous PEA levels can be achieved by inhibiting N-acylethanolamine acid amidase (NAAA), one of its primary hydrolyzing enzymes. This study aimed to test the hypothesis that NAAA inhibition, with the novel brain permeable NAAA inhibitor AM11095, modulates morphine's antinociceptive effects and attenuates the development of morphine tolerance in rats. We tested this hypothesis by measuring the pain threshold to noxious mechanical stimuli and, as a neural correlate, we conducted in vivo electrophysiological recordings from pain-sensitive locus coeruleus (LC) noradrenergic neurons in anesthetized rats. AM11095 dose-dependently (3-30 mg/kg) enhanced the antinociceptive effects of morphine and delayed the development of tolerance to chronic morphine in behaving rats. Consistently, AM11095 enhanced morphine-induced attenuation of the response of LC neurons to foot-shocks and prevented the attenuation of morphine effects following chronic treatment. Behavioral and electrophysiological effects of AM11095 on chronic morphine were paralleled by a decrease in glial activation in the spinal cord, an index of opioid-induced neuroinflammation. NAAA inhibition might represent a potential novel therapeutic approach to increase the analgesic effects of opioids and delay the development of tolerance.
Collapse
Affiliation(s)
- Mauro Congiu
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Laura Micheli
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba, Università Degli Studi Di Firenze, Florence, Italy
| | - Michele Santoni
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Claudia Sagheddu
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Anna Lisa Muntoni
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Alexandros Makriyannis
- Department of Pharmaceutical Sciences, Department of Chemistry and Chemical Biology, Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Michael S Malamas
- Department of Pharmaceutical Sciences, Department of Chemistry and Chemical Biology, Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Carla Ghelardini
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba, Università Degli Studi Di Firenze, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba, Università Degli Studi Di Firenze, Florence, Italy
| | - Marco Pistis
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
11
|
Mabou Tagne A, Fotio Y, Ibne Rashid T, Piomelli D. Persistent Exposure to Δ 9-Tetrahydrocannabinol during Adolescence Does Not Affect Nociceptive Responding in Adult Mice. J Pharmacol Exp Ther 2021; 378:215-221. [PMID: 34183435 PMCID: PMC11046735 DOI: 10.1124/jpet.121.000740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/23/2021] [Indexed: 12/28/2022] Open
Abstract
Evidence suggests that Δ9-tetrahydrocannabinol (Δ9-THC), the intoxicating component of cannabis, causes enduring changes in the structure and function of adolescent brain circuits implicated in nociceptive responding. However, whether such changes might persistently disrupt nociceptive behaviors remains unknown. In the present study, we subjected C57BL6/J mice of both sexes to once-daily injections of Δ9-THC (5 mg-kg-1, i.p.) or vehicle throughout adolescence (PND 30-43) and, when the animals had reached adulthood (PND 70), assessed nociceptive behavior using the formalin and chronic constriction injury tests. We also investigated, using the tail immersion test, the antinociceptive effects of morphine and the development of tolerance to such effects. The results show that adolescent Δ9-THC exposure does not significantly impair nociceptive responding or morphine-related antinociception and tolerance. The findings suggest that frequent exposure to a moderate dose of Δ9-THC during adolescence does not permanently alter nociceptive circuits in male or female mice. SIGNIFICANCE STATEMENT: The endocannabinoid system serves critical functions in the central and peripheral nervous systems, including regulation of pain, and can be modified by prolonged exposure to the intoxicating constituent of cannabis, Δ9-tetrahydrocannabinol (Δ9-THC). This raises the possibility that regular use of Δ9-THC-containing cannabis during adolescence might cause changes in nociception that persist into adulthood. This study found that frequent early-life exposure to a moderate dose of Δ9-THC does not permanently alter nociceptive function in male or female mice.
Collapse
Affiliation(s)
- Alex Mabou Tagne
- Department of Anatomy and Neurobiology (A.M.T., Y.F., T.I.R., D.P.), Department of Biological Chemistry, (D.P.), and Department of Pharmaceutical Sciences (D.P.), University of California, Irvine, California
| | - Yannick Fotio
- Department of Anatomy and Neurobiology (A.M.T., Y.F., T.I.R., D.P.), Department of Biological Chemistry, (D.P.), and Department of Pharmaceutical Sciences (D.P.), University of California, Irvine, California
| | - Tarif Ibne Rashid
- Department of Anatomy and Neurobiology (A.M.T., Y.F., T.I.R., D.P.), Department of Biological Chemistry, (D.P.), and Department of Pharmaceutical Sciences (D.P.), University of California, Irvine, California
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology (A.M.T., Y.F., T.I.R., D.P.), Department of Biological Chemistry, (D.P.), and Department of Pharmaceutical Sciences (D.P.), University of California, Irvine, California
| |
Collapse
|
12
|
Rock EM, Limebeer CL, Sullivan MT, DeVuono MV, Lichtman AH, Di Marzo V, Mechoulam R, Parker LA. N-Oleoylglycine and N-Oleoylalanine Do Not Modify Tolerance to Nociception, Hyperthermia, and Suppression of Activity Produced by Morphine. Front Synaptic Neurosci 2021; 13:620145. [PMID: 33767617 PMCID: PMC7985545 DOI: 10.3389/fnsyn.2021.620145] [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: 10/22/2020] [Accepted: 02/17/2021] [Indexed: 11/21/2022] Open
Abstract
The endogenous amide N-Oleoylglycine (OlGly) and its analog N-Oleoylalanine (OlAla), have been shown to interfere with the affective and somatic responses to acute naloxone-precipitated MWD in male rats. Here we evaluated the potential of a single dose (5 mg/kg, ip) which alleviates withdrawal of these endogenous fatty acid amides to modify tolerance to anti-nociception, hyperthermia, and suppression of locomotion produced by morphine in male Sprague-Dawley rats. Although rats did develop tolerance to the hypolocomotor and analgesic effects of morphine, they did not develop tolerance to the hyperthermic effects of this substance. Administration of neither OlGly nor OlAla interfered with the establishment of morphine tolerance, nor did they modify behavioral responses elicited by morphine on any trial. These results suggest that the effects of OlGly and OlAla on opiate dependence may be limited to naloxone-precipitated withdrawal effects.
Collapse
Affiliation(s)
- Erin M Rock
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Cheryl L Limebeer
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Megan T Sullivan
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Marieka V DeVuono
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Richerche, Naples, Italy.,Canada Excellence Research Chair on the Gut Microbiome/Endocannabinoidome Axis in Metabolic Health, Faculty of Medicine and Faculty of Agriculture and Food Science, CRIYUCPQ, INAF and Centre NUTRISS, Université Laval, Quebec City, QC, Canada
| | - Raphael Mechoulam
- Medical Faculty, Institute for Drug Research, Hebrew University, Jerusalem, Israel
| | - Linda A Parker
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
13
|
Lago-Fernandez A, Zarzo-Arias S, Jagerovic N, Morales P. Relevance of Peroxisome Proliferator Activated Receptors in Multitarget Paradigm Associated with the Endocannabinoid System. Int J Mol Sci 2021; 22:1001. [PMID: 33498245 PMCID: PMC7863932 DOI: 10.3390/ijms22031001] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 02/06/2023] Open
Abstract
Cannabinoids have shown to exert their therapeutic actions through a variety of targets. These include not only the canonical cannabinoid receptors CB1R and CB2R but also related orphan G protein-coupled receptors (GPCRs), ligand-gated ion channels, transient receptor potential (TRP) channels, metabolic enzymes, and nuclear receptors. In this review, we aim to summarize reported compounds exhibiting their therapeutic effects upon the modulation of CB1R and/or CB2R and the nuclear peroxisome proliferator-activated receptors (PPARs). Concomitant actions at CBRs and PPARα or PPARγ subtypes have shown to mediate antiobesity, analgesic, antitumoral, or neuroprotective properties of a variety of phytogenic, endogenous, and synthetic cannabinoids. The relevance of this multitargeting mechanism of action has been analyzed in the context of diverse pathologies. Synergistic effects triggered by combinatorial treatment with ligands that modulate the aforementioned targets have also been considered. This literature overview provides structural and pharmacological insights for the further development of dual cannabinoids for specific disorders.
Collapse
Affiliation(s)
| | | | - Nadine Jagerovic
- Medicinal Chemistry Institute, Spanish Research Council, Juan de la Cierva 3, 28006 Madrid, Spain; (A.L.-F.); (S.Z.-A.)
| | - Paula Morales
- Medicinal Chemistry Institute, Spanish Research Council, Juan de la Cierva 3, 28006 Madrid, Spain; (A.L.-F.); (S.Z.-A.)
| |
Collapse
|
14
|
Ma C, Zhang M, Liu L, Zhang P, Liu D, Zheng X, Zhong X, Wang G. Low-dose cannabinoid receptor 2 agonist induces microglial activation in a cancer pain-morphine tolerance rat model. Life Sci 2021; 264:118635. [PMID: 33131746 DOI: 10.1016/j.lfs.2020.118635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/11/2020] [Accepted: 10/18/2020] [Indexed: 12/15/2022]
Abstract
AIMS Cancer pain seriously affects the life quality of patients. Morphine is commonly used for cancer pain, but tolerance development limits its clinical administration. Central immune signaling is important in the development of cancer pain and morphine tolerance. Cannabinoid receptor 2 (CB2) inhibits cancer pain and morphine tolerance by regulating central immune signaling. In the present study, we investigated the mechanisms of central immune signaling involved in morphine tolerance inhibition by the CB2 agonist AM1241 in cancer pain treatment. MAIN METHODS Rats were implanted with tumor cells and divided into 4 groups: Vehicle (PBS), 0.07 μg AM1241, 0.03 μg AM1241, and AM630 (10 μg) + AM1241 (0.07 μg). All groups received morphine (20 μg/day, i.t.) for 8 days. AM630 (CB2 antagonist) was intrathecally injected 30 min before AM1241, and AM1241 was intrathecally injected 30 min before morphine. The spinal cord (SC) and dorsal root ganglion (DRG) were collected to determine the expression of Toll-like receptor 4 (TLR4), the p38 mitogen-activated protein kinase (MAPK), microglial markers, interleukin (IL)-1β, and tumor necrosis factor (TNF)-α. KEY FINDINGS The expression of TLR4, p38 MAPK, microglial markers, IL-1β, and TNF-α was significantly higher in AM1241-pretreated groups than in the vehicle group (P < 0.05). No difference in microglial markers, IL-1β, and TNF-α expression was detected in the AM630 + AM1241 group compared with the vehicle group. SIGNIFICANCE Our results suggest that in a cancer pain-morphine tolerance model, an i.t. non-analgesic dose of AM1241 induces microglial activation and IL-1β TNF-α upregulation in SC and DRG via the CB2 receptor pathway.
Collapse
Affiliation(s)
- Chao Ma
- Department of Anesthesiology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Mingyue Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Li Liu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Pinyi Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Dandan Liu
- Department of Anesthesiology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyu Zheng
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xuelai Zhong
- Department of Anesthesiology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Guonian Wang
- Department of Anesthesiology, The Fourth Hospital of Harbin Medical University, Harbin, China; Pain Research Institute of Heilongjiang Academy of Medical Sciences, Harbin, China.
| |
Collapse
|
15
|
Asadi Akbarabadi E, Rajabi Vardanjani H, Molavinia S, Pashmforoosh M, Khodayar MJ. PMSF Attenuates Morphine Antinociceptive Tolerance and Dependence in Mice: Its Association with the Oxidative Stress Suppression. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:300-309. [PMID: 34903990 PMCID: PMC8653646 DOI: 10.22037/ijpr.2020.112936.14038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Opioids use has been limited due to tolerance and dependence as major unwanted effects. Previous evidence has shown that targeting endocannabinoid signaling can prevent the development of opioid tolerance and dependence. This study was designed to evaluate the effect of phenylmethylsulfonyl fluoride (PMSF), an inhibitor of fatty acid amide hydrolase (FAAH), on morphine antinociceptive tolerance and physical dependence in mice. The antinociceptive effects of PMSF at the doses 60, 120, and 300 mg/kg were investigated. Results showed that PMSF has a notable antinociceptive effect at doses 120 and 300 mg/kg. The dose of (60 mg/kg, i.p.) PMSF was considered as a sub-antinociceptive dose. Morphine tolerance and dependence were induced by twice-daily injection of morphine (10 mg/kg, s.c.) for 10 consecutive days and the last dose on day 11. Tolerance was assessed by the hot-plate test and dependence by naloxone-precipitated morphine withdrawal signs. In the brain, oxidative stress markers include activities of glutathione peroxidase, catalase, superoxide dismutase, and levels of malondialdehyde and glutathione were determined. A sub-antinociceptive dose (60 mg/kg) of PMSF could reduce tolerance in both acute and chronic methods of administration. However, alleviation of dependence and suppression of oxidative stress markers occurred in the chronic administration of PMSF. In conclusion, it seems that PMSF can suppress morphine tolerance and dependence. However, more studies are needed to clarify its mechanism.
Collapse
Affiliation(s)
- Ehsan Asadi Akbarabadi
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Hossein Rajabi Vardanjani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Shahrzad Molavinia
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. ,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | | | - Mohammad Javad Khodayar
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. ,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. ,Corresponding author: E-mail:
| |
Collapse
|
16
|
Slivicki RA, Iyer V, Mali SS, Garai S, Thakur GA, Crystal JD, Hohmann AG. Positive Allosteric Modulation of CB 1 Cannabinoid Receptor Signaling Enhances Morphine Antinociception and Attenuates Morphine Tolerance Without Enhancing Morphine- Induced Dependence or Reward. Front Mol Neurosci 2020; 13:54. [PMID: 32410959 PMCID: PMC7199816 DOI: 10.3389/fnmol.2020.00054] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/17/2020] [Indexed: 01/09/2023] Open
Abstract
Opioid analgesics represent a critical treatment for chronic pain in the analgesic ladder of the World Health Organization. However, their use can result in a number of unwanted side-effects including incomplete efficacy, constipation, physical dependence, and overdose liability. Cannabinoids enhance the pain-relieving effects of opioids in preclinical studies and dampen unwanted side-effects resulting from excessive opioid intake. We recently reported that a CB1 positive allosteric modulator (PAM) exhibits antinociceptive efficacy in models of pathological pain and lacks the adverse side effects of direct CB1 receptor activation. In the present study, we evaluated whether a CB1 PAM would enhance morphine’s therapeutic efficacy in an animal model of chemotherapy-induced neuropathic pain and characterized its impact on unwanted side-effects associated with chronic opioid administration. In paclitaxel-treated mice, both the CB1 PAM GAT211 and the opioid analgesic morphine reduced paclitaxel-induced behavioral hypersensitivities to mechanical and cold stimulation in a dose-dependent manner. Isobolographic analysis revealed that combinations of GAT211 and morphine resulted in anti-allodynic synergism. In paclitaxel-treated mice, a sub-threshold dose of GAT211 prevented the development of tolerance to the anti-allodynic effects of morphine over 20 days of once daily dosing. However, GAT211 did not reliably alter somatic withdrawal signs (i.e., jumps, paw tremors) in morphine-dependent neuropathic mice challenged with naloxone. In otherwise naïve mice, GAT211 also prolonged antinociceptive efficacy of morphine in the tail-flick test and reduced the overall right-ward shift in the ED50 for morphine to produce antinociception in the tail-flick test, consistent with attenuation of morphine tolerance. Pretreatment with GAT211 did not alter somatic signs of μ opioid receptor dependence in mice rendered dependent upon morphine via subcutaneous implantation of a morphine pellet. Moreover, GAT211 did not reliably alter μ-opioid receptor-mediated reward as measured by conditioned place preference to morphine. Our results suggest that a CB1 PAM may be beneficial in enhancing and prolonging the therapeutic properties of opioids while potentially sparing unwanted side-effects (e.g., tolerance) that occur with repeated opioid treatment.
Collapse
Affiliation(s)
- Richard A Slivicki
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Vishakh Iyer
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Sonali S Mali
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Sumanta Garai
- Center for Drug Discovery, Bouve College of Health Sciences, Northeastern University, Boston, MA, United States
| | - Ganesh A Thakur
- Center for Drug Discovery, Bouve College of Health Sciences, Northeastern University, Boston, MA, United States
| | - Jonathon D Crystal
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Andrea G Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States.,Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States
| |
Collapse
|
17
|
Fotio Y, Palese F, Guaman Tipan P, Ahmed F, Piomelli D. Inhibition of fatty acid amide hydrolase in the CNS prevents and reverses morphine tolerance in male and female mice. Br J Pharmacol 2020; 177:3024-3035. [PMID: 32077093 DOI: 10.1111/bph.15031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Fatty acid amide hydrolase (FAAH) is an intracellular serine amidase that terminates the signalling of various lipid messengers involved in pain regulation, including anandamide and palmitoylethanolamide. Here, we investigated the effects of pharmacological or genetic FAAH removal on tolerance to the anti-nociceptive effects of morphine. EXPERIMENTAL APPROACH We induced tolerance in male and female mice by administering twice-daily morphine for 7 days while monitoring nociceptive thresholds by the tail immersion test. The globally active FAAH inhibitor URB597 (1 and 3 mg·kg-1 , i.p.) or the peripherally restricted FAAH inhibitor URB937 (3 mg·kg-1 , i.p.) were administered daily 30 min prior to morphine, alone or in combination with the cannabinoid CB1 receptor antagonist AM251 (3 mg·kg-1 , i.p.), the CB2 receptor antagonist AM630 (3 mg·kg-1 , i.p.), or the PPAR-α antagonist GW6471 (4 mg·kg-1 , i.p.). Spinal levels of FAAH-regulated lipids were quantified by LC/MS-MS. Gene transcription was assessed by RT-qPCR. KEY RESULTS URB597 prevented and reversed morphine tolerance in both male and female mice. This effect was mimicked by genetic FAAH deletion, but not by URB937. Treatment with AM630 suppressed, whereas treatment with AM251 or GW6471, attenuated the effects of URB597. Anandamide mobilization was enhanced in the spinal cord of morphine-tolerant mice. mRNA levels of the anandamide-producing enzyme N-acyl-phosphatidylethanolamine PLD (NAPE-PLD) and the palmitoylethanolamide receptor PPAR-α, but not those for CB2 , CB1 receptors or FAAH, were elevated in spinal cord CONCLUSION AND IMPLICATIONS: FAAH-regulated lipid signalling in the CNS modulated opiate tolerance, suggesting FAAH as a potential target for opiate-sparing medications.
Collapse
Affiliation(s)
- Yannick Fotio
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, California
| | - Francesca Palese
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, California
| | - Pablo Guaman Tipan
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, California
| | - Faizy Ahmed
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, California.,Center for the Study of Cannabis, University of California, Irvine, Irvine, California
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, California.,Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California.,Center for the Study of Cannabis, University of California, Irvine, Irvine, California
| |
Collapse
|