1
|
Gamble MC, Williams BR, Singh N, Posa L, Freyberg Z, Logan RW, Puig S. Mu-opioid receptor and receptor tyrosine kinase crosstalk: Implications in mechanisms of opioid tolerance, reduced analgesia to neuropathic pain, dependence, and reward. Front Syst Neurosci 2022; 16:1059089. [PMID: 36532632 PMCID: PMC9751598 DOI: 10.3389/fnsys.2022.1059089] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/31/2022] [Indexed: 07/30/2023] Open
Abstract
Despite the prevalence of opioid misuse, opioids remain the frontline treatment regimen for severe pain. However, opioid safety is hampered by side-effects such as analgesic tolerance, reduced analgesia to neuropathic pain, physical dependence, or reward. These side effects promote development of opioid use disorders and ultimately cause overdose deaths due to opioid-induced respiratory depression. The intertwined nature of signaling via μ-opioid receptors (MOR), the primary target of prescription opioids, with signaling pathways responsible for opioid side-effects presents important challenges. Therefore, a critical objective is to uncouple cellular and molecular mechanisms that selectively modulate analgesia from those that mediate side-effects. One such mechanism could be the transactivation of receptor tyrosine kinases (RTKs) via MOR. Notably, MOR-mediated side-effects can be uncoupled from analgesia signaling via targeting RTK family receptors, highlighting physiological relevance of MOR-RTKs crosstalk. This review focuses on the current state of knowledge surrounding the basic pharmacology of RTKs and bidirectional regulation of MOR signaling, as well as how MOR-RTK signaling may modulate undesirable effects of chronic opioid use, including opioid analgesic tolerance, reduced analgesia to neuropathic pain, physical dependence, and reward. Further research is needed to better understand RTK-MOR transactivation signaling pathways, and to determine if RTKs are a plausible therapeutic target for mitigating opioid side effects.
Collapse
Affiliation(s)
- Mackenzie C. Gamble
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Molecular and Translational Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Benjamin R. Williams
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Navsharan Singh
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Luca Posa
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ryan W. Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
| | - Stephanie Puig
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
2
|
Puig S, Shelton MA, Barko K, Seney ML, Logan RW. Sex-specific role of the circadian transcription factor NPAS2 in opioid tolerance, withdrawal and analgesia. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12829. [PMID: 36053258 PMCID: PMC9744556 DOI: 10.1111/gbb.12829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/09/2022] [Accepted: 08/09/2022] [Indexed: 02/05/2023]
Abstract
Opioids like fentanyl remain the mainstay treatment for chronic pain. Unfortunately, opioid's high dependence liability has led to the current opioid crisis, in part, because of side-effects that develop during long-term use, including analgesic tolerance and physical dependence. Both tolerance and dependence to opioids may lead to escalation of required doses to achieve previous therapeutic efficacy. Additionally, altered sleep and circadian rhythms are common in people on opioid therapy. Opioids impact sleep and circadian rhythms, while disruptions to sleep and circadian rhythms likely mediate the effects of opioids. However, the mechanisms underlying these bidirectional relationships between circadian rhythms and opioids remain largely unknown. The circadian protein, neuronal PAS domain protein 2 (NPAS2), regulates circadian-dependent gene transcription in structure of the central nervous system that modulate opioids and pain. Here, male and female wild-type and NPAS2-deficient (NPAS2-/-) mice were used to investigate the role of NPAS2 in fentanyl analgesia, tolerance, hyperalgesia and physical dependence. Overall, thermal pain thresholds, acute analgesia and tolerance to a fixed dose of fentanyl were largely similar between wild-type and NPAS2-/- mice. However, female NPAS2-/- exhibited augmented analgesic tolerance and significantly more behavioral symptoms of physical dependence to fentanyl. Only male NPAS2-/- mice had increased fentanyl-induced hypersensitivity, when compared with wild-type males. Together, our findings suggest sex-specific effects of NPAS2 signaling in the regulation of fentanyl-induced tolerance, hyperalgesia and dependence.
Collapse
Affiliation(s)
- Stephanie Puig
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
- Translational Neuroscience Program, Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Micah A. Shelton
- Translational Neuroscience Program, Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Kelly Barko
- Translational Neuroscience Program, Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Marianne L. Seney
- Translational Neuroscience Program, Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Ryan W. Logan
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
- Translational Neuroscience Program, Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Center for Systems NeuroscienceBoston UniversityBostonMassachusettsUSA
| |
Collapse
|
3
|
Inhibiting Hv1 channel in peripheral sensory neurons attenuates chronic inflammatory pain and opioid side effects. Cell Res 2022; 32:461-476. [PMID: 35115667 PMCID: PMC9061814 DOI: 10.1038/s41422-022-00616-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 01/04/2022] [Indexed: 02/05/2023] Open
Abstract
Both opioids and nonsteroidal anti-inflammatory drugs (NSAIDS) produce deleterious side effects and fail to provide sustained relief in patients with chronic inflammatory pain. Peripheral neuroinflammation (PN) is critical for initiation and development of inflammatory pain. A better understanding of molecular mechanisms underlying PN would facilitate the discovery of new analgesic targets and the development of new therapeutics. Emerging evidence suggests that peripheral sensory neurons are not only responders to painful stimuli, but are also actively engaged in inflammation and immunity, whereas the intrinsic regulatory mechanism is poorly understood. Here we report the expression of proton-selective ion channel Hv1 in peripheral sensory neurons in rodents and humans, which was previously shown as selectively expressed in microglia in mammalian central nervous system. Neuronal Hv1 was up-regulated by PN or depolarizing stimulation, which in turn aggravates inflammation and nociception. Inhibiting neuronal Hv1 genetically or by a newly discovered selective inhibitor YHV98-4 reduced intracellular alkalization and ROS production in inflammatory pain, mitigated the imbalance in downstream SHP-1-pAKT signaling, and also diminished pro-inflammatory chemokine release to alleviate nociception and morphine-induced hyperalgesia and tolerance. Thus, our data reveal neuronal Hv1 as a novel target in analgesia strategy and managing opioids-related side effects.
Collapse
|
4
|
Scholz O, Otter S, Welters A, Wörmeyer L, Dolenšek J, Klemen MS, Pohorec V, Eberhard D, Mrugala J, Hamacher A, Koch A, Sanz M, Hoffmann T, Hogeback J, Herebian D, Klöcker N, Piechot A, Mayatepek E, Meissner T, Stožer A, Lammert E. Peripherally active dextromethorphan derivatives lower blood glucose levels by targeting pancreatic islets. Cell Chem Biol 2021; 28:1474-1488.e7. [PMID: 34118188 DOI: 10.1016/j.chembiol.2021.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 03/09/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022]
Abstract
Dextromethorphan (DXM) acts as cough suppressant via its central action. Cell-protective effects of this drug have been reported in peripheral tissues, making DXM potentially useful for treatment of several common human diseases, such as type 2 diabetes mellitus (T2DM). Pancreatic islets are among the peripheral tissues that positively respond to DXM, and anti-diabetic effects of DXM were observed in two placebo-controlled, randomized clinical trials in humans with T2DM. Since these effects were associated with central side effects, we here developed chemical derivatives of DXM that pass the blood-brain barrier to a significantly lower extent than the original drug. We show that basic nitrogen-containing residues block central adverse events of DXM without reducing its anti-diabetic effects, including the protection of human pancreatic islets from cell death. These results show how to chemically modify DXM, and possibly other morphinans, as to exclude central side effects, while targeting peripheral tissues, such as pancreatic islets.
Collapse
Affiliation(s)
- Okka Scholz
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Silke Otter
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany
| | - Alena Welters
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Laura Wörmeyer
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Jurij Dolenšek
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia
| | - Maša Skelin Klemen
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Viljem Pohorec
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Daniel Eberhard
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Jessica Mrugala
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Anna Hamacher
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Angela Koch
- Institute of Neuro- and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Miguel Sanz
- Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; Taros Chemicals GmbH & Co. KG, 44227 Dortmund, Germany
| | - Torsten Hoffmann
- Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; Taros Chemicals GmbH & Co. KG, 44227 Dortmund, Germany
| | - Jens Hogeback
- A&M Labor für Analytik und Metabolismusforschung Service GmbH, 50126 Bergheim, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Nikolaj Klöcker
- Institute of Neuro- and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Alexander Piechot
- Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; Taros Chemicals GmbH & Co. KG, 44227 Dortmund, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Eckhard Lammert
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
| |
Collapse
|
5
|
Zhang L, Roy S. Opioid Modulation of the Gut-Brain Axis in Opioid-Associated Comorbidities. Cold Spring Harb Perspect Med 2021; 11:a040485. [PMID: 32816876 PMCID: PMC8415294 DOI: 10.1101/cshperspect.a040485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Growing evidence from animal and human studies show that opioids have a major impact on the composition and function of gut microbiota. This leads to disruption in gut permeability and altered microbial metabolites, driving both systemic and neuroinflammation, which in turn impacts central nervous system (CNS) homeostasis. Tolerance and dependence are the major comorbidities associated with prolonged opioid use. Inflammatory mediators and signaling pathways have been implicated in both opioid tolerance and dependence. We provide evidence that targeting the gut microbiome during opioid use through prebiotics, probiotics, antibiotics, and fecal microbial transplantation holds the greatest promise for novel treatments for opioid abuse. Basic research and clinical trials are required to examine what is more efficacious to yield new insights into the role of the gut-brain axis in opioid abuse.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmacology, University of Minnesota, Minnesota McGuire Translational Research Facility, Minneapolis, Minnesota 55455, USA
| | - Sabita Roy
- Department of Pharmacology, University of Minnesota, Minnesota McGuire Translational Research Facility, Minneapolis, Minnesota 55455, USA
- Department of Surgery, University of Miami, Miami, Florida 33153, USA
| |
Collapse
|
6
|
Lin M, Deng K, Li Y, Wan J. Morphine enhances LPS-induced macrophage apoptosis through a PPARγ-dependent mechanism. Exp Ther Med 2021; 22:714. [PMID: 34007323 PMCID: PMC8120503 DOI: 10.3892/etm.2021.10146] [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: 10/12/2020] [Accepted: 03/29/2021] [Indexed: 12/17/2022] Open
Abstract
Morphine has been widely used for the treatment of pain and extensive studies have revealed a regulatory role for morphine in cell apoptosis. However, the molecular mechanisms underlying morphine-mediated apoptosis remain to be fully elucidated. The present study aimed to investigate the effects of morphine on lipopolysaccharide (LPS)-induced bone marrow-derived macrophage (BMDM) apoptosis and to determine the role of the peroxisome proliferator-activated receptor (PPAR)γ signaling pathway in this process. BMDMs were isolated from BALB/c mice and stimulated with LPS. Hoechst 33342 staining and flow cytometric analysis were performed to evaluate the effects of morphine on LPS-induced apoptosis of BMDMs. Caspase activity assays were used to determine the involvement of the apoptosis pathway. The expression levels of caspase-3, caspase-8, caspase-9 and PPARγ were analyzed using western blotting. Finally, GW9662, a specific PPARγ antagonist, was used to determine whether the regulatory effects of morphine on LPS-induced BMDM apoptosis were PPARγ-dependent. The results of the present study revealed that morphine increased the apoptosis of LPS-stimulated BMDMs. Morphine upregulated the expression levels and activity of caspase-3 in LPS-stimulated BMDMs, but downregulated the expression levels and activity of caspase-8. Morphine treatment also upregulated LPS-induced PPARγ expression levels in BMDMs. Finally, the stimulatory effects of morphine on LPS-induced apoptosis and caspase-3/9 activation were markedly reduced by GW9662. In conclusion, the findings of the present study indicated that morphine significantly promoted LPS-induced BMDM apoptosis and caspase-3/9 activation. These results suggested that the intrinsic pathway of apoptosis may be involved in the proapoptotic effects of morphine on LPS-stimulated BMDMs, which may be dependent, at least partially, on PPARγ activation.
Collapse
Affiliation(s)
- Mingying Lin
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Keqiong Deng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ya Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
7
|
Rieb LM, DeBeck K, Hayashi K, Wood E, Nosova E, Milloy MJ. Withdrawal-associated injury site pain prevalence and correlates among opioid-using people who inject drugs in Vancouver, Canada. Drug Alcohol Depend 2020; 216:108242. [PMID: 32861135 PMCID: PMC7850369 DOI: 10.1016/j.drugalcdep.2020.108242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Pain can return temporarily to old injury sites during opioid withdrawal. The prevalence and impact of opioid withdrawal-associated injury site pain (WISP) in various groups is unknown. METHODS Using data from observational cohorts, we estimated the prevalence and correlates of WISP among opioid-using people who inject drugs (PWID). Between June and December 2015, data on WISP and opioid use behaviours were elicited from participants in three ongoing prospective cohort studies in Vancouver, Canada, who were aged 18 years and older and who self-reported at least daily injection of heroin or non-medical presciption opioids. RESULTS Among 631 individuals, 276 (43.7 %) had a healed injury (usually pain-free), among whom 112 (40.6 %) experienced WISP, representing 17.7 % of opioid-using PWID interviewed. In a multivariable logistic regression model, WISP was positively associated with having a high school diploma or above (Adjusted Odds Ratio [AOR] = 2.23, 95 % Confidence Interval [CI]: 1.31-3.84), any heroin use in the last six months (AOR = 2.00, 95 % CI: 1.14-3.57), feeling daily pain that required medication (AOR = 2.06, CI: 1.18-3.63), and negatively associated with older age at first drug use (AOR = 0.96, 95 % CI: 0.93-0.99). Among 112 individuals with WISP, 79 (70.5 %) said that having this pain affected their opioid use behaviour, of whom 57 (72.2 %) used more opioids, 19 (24.1 %) avoided opioid withdrawal, while 3 (3.8 %) no longer used opioids to avoid WISP. CONCLUSIONS WISP is prevalent among PWID with a previous injury, and may alter opioid use patterns. Improved care strategies for WISP are warrented.
Collapse
Affiliation(s)
- Launette Marie Rieb
- Department of Family Practice, University of British Columbia, 5950 University Boulevard, Vancouver, British Columbia, Canada
| | - Kora DeBeck
- School of Public Policy, Simon Fraser University, 515 West Hastings Street, Office 3269, Vancouver, British Columbia, Canada,British Columbia Centre on Substance Use, 1045 Howe Street, Vancouver, British Columbia, Canada
| | - Kanna Hayashi
- British Columbia Centre on Substance Use, 1045 Howe Street, Vancouver, British Columbia, Canada,Faculty of Health Sciences, Simon Fraser University, Blusson Hall, Room 11300, Burnaby, British Columbia, Canada
| | - Evan Wood
- British Columbia Centre on Substance Use, 1045 Howe Street, Vancouver, British Columbia, Canada,Department of Medicine, University of British Columbia, 2775 Laurel Street, 10thFloor, Vancouver, British Columbia, Canada
| | - Ekaterina Nosova
- British Columbia Centre on Substance Use, 1045 Howe Street, Vancouver, British Columbia, Canada
| | - M-J Milloy
- British Columbia Centre on Substance Use, 1045 Howe Street, Vancouver, British Columbia, Canada; Department of Medicine, University of British Columbia, 2775 Laurel Street, 10thFloor, Vancouver, British Columbia, Canada.
| |
Collapse
|
8
|
Jacob JC, Sakakibara K, Mischel RA, Henderson G, Dewey WL, Akbarali HI. Ethanol Reversal of Oxycodone Tolerance in Dorsal Root Ganglia Neurons. Mol Pharmacol 2018; 93:417-426. [PMID: 29467238 DOI: 10.1124/mol.117.110775] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 02/08/2018] [Indexed: 01/23/2023] Open
Abstract
Oxycodone is a semisynthetic opioid compound that is widely prescribed, used, and abused today, and has a well-established role in shaping the current opioid epidemic. Previously, we have shown that tolerance develops to the antinociceptive and respiratory depressive effects of oxycodone in mice, and that a moderate dose of acute ethanol or a protein kinase C (PKC) inhibitor reversed that tolerance. To investigate further if tolerance was occurring through neuronal mechanisms, our aims for this study were to assess the effects of acute and prolonged oxycodone in isolated dorsal root ganglia (DRG) neurons and to determine if this tolerance was reversed by either ethanol or a PKC inhibitor. We found that an acute exposure to 3 μM oxycodone reduced neuronal excitability, as measured by increased threshold potentials and reduced action potential amplitude, without eliciting measurable changes in resting membrane potential. Exposure to 10 μM oxycodone for 18-24 hours prevented oxycodone's effect on neuronal excitability, indicative of tolerance development. The development of opioid tolerance was mitigated in DRG neurons from β-arrestin 2 knockout mice. Oxycodone tolerance was reversed in isolated DRG neurons by the acute application of either ethanol (20 mM) or the PKC inhibitor, bisindolylmaleimide XI hydrochloride (Bis XI), when a challenge of 3 µM oxycodone significantly reduced neuronal excitability following prolonged exposure. Through these studies, we concluded that oxycodone acutely reduced neuronal excitability, tolerance developed to this effect, and reversal of that tolerance occurred at the level of a single neuron, suggesting that reversal of oxycodone tolerance by either ethanol or Bis XI involves cellular mechanisms.
Collapse
Affiliation(s)
- Joanna C Jacob
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.C.J., K.S., R.A.M., W.L.D., H.I.A.); and School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (G.H.)
| | - Kensuke Sakakibara
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.C.J., K.S., R.A.M., W.L.D., H.I.A.); and School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (G.H.)
| | - Ryan A Mischel
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.C.J., K.S., R.A.M., W.L.D., H.I.A.); and School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (G.H.)
| | - Graeme Henderson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.C.J., K.S., R.A.M., W.L.D., H.I.A.); and School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (G.H.)
| | - William L Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.C.J., K.S., R.A.M., W.L.D., H.I.A.); and School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (G.H.)
| | - Hamid I Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.C.J., K.S., R.A.M., W.L.D., H.I.A.); and School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (G.H.)
| |
Collapse
|
9
|
Wong W. New connections: Getting the good without the bad, opiate edition. Sci Signal 2017; 10:10/471/eaan2516. [DOI: 10.1126/scisignal.aan2516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Researchers identify strategies to limit the adverse effects and liabilities of opioid receptor–targeting drugs.
Collapse
Affiliation(s)
- Wei Wong
- Science Signaling, AAAS, Washington, DC 20005, USA
| |
Collapse
|