1
|
Gooding SW, Felth L, Foxall R, Rosa Z, Ireton K, Sall I, Gipoor J, Gaur A, King M, Dirks N, Whistler CA, Whistler JL. Deletion of arrestin-3 does not reduce drug-seeking behavior in a longitudinal paradigm of oral morphine self-administration. Front Pharmacol 2024; 15:1438037. [PMID: 39391692 PMCID: PMC11464476 DOI: 10.3389/fphar.2024.1438037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Opioid drugs are potent analgesics that mimic the endogenous opioid peptides, endorphins and enkephalins, by activating the µ-opioid receptor. Opioid use is limited by side effects, including significant risk of opioid use disorder. Improvement of the effect/side effect profile of opioid medications is a key pursuit of opioid research, yet there is no consensus on how to achieve this goal. One hypothesis is that the degree of arrestin-3 recruitment to the µ-opioid receptor impacts therapeutic utility. However, it is not clear whether increased or decreased interaction of the µ-opioid receptor with arrestin-3 would reduce compulsive drug-seeking. Methods We utilized three genotypes of mice with varying abilities to recruit arrestin-3 to the µ-opioid receptor in response to morphine in a novel longitudinal operant self-administration model. We also created a quantitative method to define compulsivity in drug-seeking based on a multi-variate analysis of several operant response variables. Results We demonstrate that arrestin-3 knockout and wild type mice have highly variable drug-seeking behavior with few genotype differences. In contrast, in mice where the µ-opioid receptor strongly recruits arrestin-3, drug-seeking behavior is much less varied. We found that mice lacking arrestin-3 were more likely to meet the criteria for compulsivity whereas mice with enhanced arrestin-3 recruitment did not develop a compulsive phenotype. Conclusion These experiments show that a lack of arrestin-3 is not protective against the abuse liability of morphine in an operant self-administration context. Our data also suggest that opioids that engage both G protein and arrestin-3, recapitulating the endogenous signaling pattern, will reduce abuse liability.
Collapse
Affiliation(s)
- Sarah Warren Gooding
- Center for Neuroscience, University of California–Davis, Davis, CA, United States
| | - Lindsey Felth
- Center for Neuroscience, University of California–Davis, Davis, CA, United States
| | - Randi Foxall
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, United States
| | - Zachary Rosa
- Center for Neuroscience, University of California–Davis, Davis, CA, United States
| | - Kyle Ireton
- Center for Neuroscience, University of California–Davis, Davis, CA, United States
| | - Izabella Sall
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, United States
| | - Joshua Gipoor
- Center for Neuroscience, University of California–Davis, Davis, CA, United States
| | - Anirudh Gaur
- Center for Neuroscience, University of California–Davis, Davis, CA, United States
| | - Madeline King
- Center for Neuroscience, University of California–Davis, Davis, CA, United States
| | - Noah Dirks
- Center for Neuroscience, University of California–Davis, Davis, CA, United States
| | - Cheryl Allyne Whistler
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, United States
| | - Jennifer Lynne Whistler
- Center for Neuroscience, University of California–Davis, Davis, CA, United States
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
2
|
Cui D, Zhang Y, Zhang M. The effect of cannabinoid type 2 receptor agonist on morphine tolerance. IBRO Neurosci Rep 2024; 16:43-50. [PMID: 38145173 PMCID: PMC10733637 DOI: 10.1016/j.ibneur.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/24/2023] [Accepted: 11/14/2023] [Indexed: 12/26/2023] Open
Abstract
Pain highly impacts the quality of life of patients. Morphine is used for pain treatment; however, its side effects, especially morphine tolerance, limit its use in the clinic. The problem of morphine tolerance has plagued health workers and patients for years. Unfortunately, the exact mechanism of morphine tolerance has not been fully clarified. The mechanisms of morphine tolerance that are currently being studied may include μ-opioid receptor (MOR) desensitization and internalization, mitogen-activated protein kinase (MAPK) pathway activation and crosstalk, the effects of microglia and the increase in inflammatory factors. Morphine tolerance can be alleviated by improving the pathophysiological changes that lead to morphine tolerance. Previous studies have shown that a cannabinoid type 2 (CB2) receptor agonist could attenuate morphine tolerance in a variety of animal models. Many studies have shown an interaction between the cannabinoid system and the opioid system. The CB2 receptor may modulate the effect of morphine through a pathway that is common to the MOR, since both receptors are G protein-coupled receptors (GPCRs). This study introduces the potential mechanism of morphine tolerance and the effect of CB2 receptor agonists on reducing morphine tolerance, which can provide new ideas for researchers studying morphine and provide beneficial effects for patients suffering from morphine tolerance.
Collapse
Affiliation(s)
- Di Cui
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuanyuan Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mingyue Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
3
|
Gooding SW, Felth L, Foxall R, Rosa Z, Ireton K, Sall I, Gipoor J, Gaur A, King M, Dirks N, Whistler CA, Whistler JL. Deletion of arrestin-3 does not improve compulsive drug-seeking behavior in a longitudinal paradigm of oral morphine self-administration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.30.534994. [PMID: 38562752 PMCID: PMC10983877 DOI: 10.1101/2023.03.30.534994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Opioid drugs are potent analgesics that mimic the endogenous opioid peptides, endorphins and enkephalins, by activating the μ-opioid receptor. Opioid use is limited by side effects, including significant risk of opioid use disorder. Improvement of the effect/side effect profile of opioid medications is a key pursuit of opioid research, yet there is no consensus on how to achieve this goal. One hypothesis is that the degree of arrestin-3 recruitment to the μ-opioid receptor impacts therapeutic utility. However, it is not clear whether increased or decreased interaction of the μ-opioid receptor with arrestin-3 would reduce compulsive drug-seeking. To examine this question, we utilized three genotypes of mice with varying abilities to recruit arrestin-3 to the μ-opioid receptor in response to morphine in a novel longitudinal operant self-administration model. We demonstrate that arrestin-3 knockout and wild type mice have highly variable drug-seeking behavior with few genotype differences. In contrast, in mice where the μ-opioid receptor strongly recruits arrestin-3, drug-seeking behavior is much less varied. We created a quantitative method to define compulsivity in drug-seeking and found that mice lacking arrestin-3 were more likely to meet the criteria for compulsivity whereas mice with enhanced arrestin-3 recruitment did not develop a compulsive phenotype. Our data suggest that opioids that engage both G protein and arrestin-3, recapitulating the endogenous signaling pattern, will reduce abuse liability.
Collapse
Affiliation(s)
| | - Lindsey Felth
- Center for Neuroscience, University of California-Davis, Davis, CA, USA
| | - Randi Foxall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire
| | - Zachary Rosa
- Center for Neuroscience, University of California-Davis, Davis, CA, USA
| | - Kyle Ireton
- Center for Neuroscience, University of California-Davis, Davis, CA, USA
| | - Izabella Sall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire
| | - Joshua Gipoor
- Center for Neuroscience, University of California-Davis, Davis, CA, USA
| | - Anirudh Gaur
- Center for Neuroscience, University of California-Davis, Davis, CA, USA
| | - Madeline King
- Center for Neuroscience, University of California-Davis, Davis, CA, USA
| | - Noah Dirks
- Center for Neuroscience, University of California-Davis, Davis, CA, USA
| | - Cheryl A Whistler
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire
| | - Jennifer L Whistler
- Center for Neuroscience, University of California-Davis, Davis, CA, USA
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
4
|
Ochandarena NE, Niehaus JK, Tassou A, Scherrer G. Cell-type specific molecular architecture for mu opioid receptor function in pain and addiction circuits. Neuropharmacology 2023; 238:109597. [PMID: 37271281 PMCID: PMC10494323 DOI: 10.1016/j.neuropharm.2023.109597] [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: 04/12/2023] [Accepted: 05/13/2023] [Indexed: 06/06/2023]
Abstract
Opioids are potent analgesics broadly used for pain management; however, they can produce dangerous side effects including addiction and respiratory depression. These harmful effects have led to an epidemic of opioid abuse and overdose deaths, creating an urgent need for the development of both safer pain medications and treatments for opioid use disorders. Both the analgesic and addictive properties of opioids are mediated by the mu opioid receptor (MOR), making resolution of the cell types and neural circuits responsible for each of the effects of opioids a critical research goal. Single-cell RNA sequencing (scRNA-seq) technology is enabling the identification of MOR-expressing cell types throughout the nervous system, creating new opportunities for mapping distinct opioid effects onto newly discovered cell types. Here, we describe molecularly defined MOR-expressing neuronal cell types throughout the peripheral and central nervous systems and their potential contributions to opioid analgesia and addiction.
Collapse
Affiliation(s)
- Nicole E Ochandarena
- Neuroscience Curriculum, Biological and Biomedical Sciences Program, The University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Jesse K Niehaus
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; New York Stem Cell Foundation - Robertson Investigator, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
5
|
Degrandmaison J, Rochon-Haché S, Parent JL, Gendron L. Knock-In Mouse Models to Investigate the Functions of Opioid Receptors in vivo. Front Cell Neurosci 2022; 16:807549. [PMID: 35173584 PMCID: PMC8841419 DOI: 10.3389/fncel.2022.807549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/04/2022] [Indexed: 12/28/2022] Open
Abstract
Due to their low expression levels, complex multi-pass transmembrane structure, and the current lack of highly specific antibodies, the assessment of endogenous G protein-coupled receptors (GPCRs) remains challenging. While most of the research regarding their functions was performed in heterologous systems overexpressing the receptor, recent advances in genetic engineering methods have allowed the generation of several unique mouse models. These animals proved to be useful to investigate numerous aspects underlying the physiological functions of GPCRs, including their endogenous expression, distribution, interactome, and trafficking processes. Given their significant pharmacological importance and central roles in the nervous system, opioid peptide receptors (OPr) are often referred to as prototypical receptors for the study of GPCR regulatory mechanisms. Although only a few GPCR knock-in mouse lines have thus far been generated, OPr are strikingly well represented with over 20 different knock-in models, more than half of which were developed within the last 5 years. In this review, we describe the arsenal of OPr (mu-, delta-, and kappa-opioid), as well as the opioid-related nociceptin/orphanin FQ (NOP) receptor knock-in mouse models that have been generated over the past years. We further highlight the invaluable contribution of such models to our understanding of the in vivo mechanisms underlying the regulation of OPr, which could be conceivably transposed to any other GPCR, as well as the limitations, future perspectives, and possibilities enabled by such tools.
Collapse
Affiliation(s)
- Jade Degrandmaison
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Médecine, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Quebec Network of Junior Pain Investigators, Sherbrooke, QC, Canada
| | - Samuel Rochon-Haché
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Médecine, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Quebec Network of Junior Pain Investigators, Sherbrooke, QC, Canada
| | - Jean-Luc Parent
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Médecine, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Jean-Luc Parent,
| | - Louis Gendron
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Quebec Pain Research Network, Sherbrooke, QC, Canada
- *Correspondence: Louis Gendron,
| |
Collapse
|
6
|
HA-MOP knockin mice express the canonical µ-opioid receptor but lack detectable splice variants. Commun Biol 2021; 4:1070. [PMID: 34522000 PMCID: PMC8440528 DOI: 10.1038/s42003-021-02580-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/17/2021] [Indexed: 12/31/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are notoriously difficult to detect in native tissues. In an effort to resolve this problem, we have developed a novel mouse model by fusing the hemagglutinin (HA)-epitope tag sequence to the amino-terminus of the µ-opioid receptor (MOP). Although HA-MOP knock-in mice exhibit reduced receptor expression, we found that this approach allowed for highly efficient immunodetection of low abundant GPCR targets. We also show that the HA-tag facilitates both high-resolution imaging and immunoisolation of MOP. Mass spectrometry (MS) confirmed post-translational modifications, most notably agonist-selective phosphorylation of carboxyl-terminal serine and threonine residues. MS also unequivocally identified the carboxyl-terminal 387LENLEAETAPLP398 motif, which is part of the canonical MOP sequence. Unexpectedly, MS analysis of brain lysates failed to detect any of the 15 MOP isoforms that have been proposed to arise from alternative splicing of the MOP carboxyl-terminus. For quantitative analysis, we performed multiple successive rounds of immunodepletion using the well-characterized rabbit monoclonal antibody UMB-3 that selectively detects the 387LENLEAETAPLP398 motif. We found that >98% of HA-tagged MOP contain the UMB-3 epitope indicating that virtually all MOP expressed in the mouse brain exhibit the canonical amino acid sequence. Fritzwanker et al. develop a knock-in transgenic mouse line in which the hemagglutinin epitope tag sequence is fused with the amino-terminus of the µ-opioid receptor. Their model enables more efficient immunodetection of G protein-coupled receptors.
Collapse
|
7
|
Camacho E, Marie N, Dupas Q, Martel C, Nowoczyn M, Elie N, Rochais C, Töth G, Allouche S. Impact of T161, Y318 and S363 alanine mutations on regulation of the human delta-opioid receptor (hDOPr) induced by peptidic and alkaloid agonists. Neuropharmacology 2020; 179:108286. [PMID: 32841607 DOI: 10.1016/j.neuropharm.2020.108286] [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: 11/12/2019] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 11/16/2022]
Abstract
Previously, we showed a differential regulation of the human delta-opioid receptor (hDOPr) by etorphine and [D-Pen2, D-Pen5] enkephalin (DPDPE). To understand the molecular basis of such differences, we introduced 3 alanine mutations at the residues T161. Y318 and S363. Both wild type (WT) and hDOPr mutants were expressed in HEK cells containing endogenous arrestins or CFP-tagged arrestin 3, then desensitization, internalization, recycling and phosphorylation were studied. In a context of endogenous arrestin expression, a major difference in DOPr desensitization was observed between agonists that was modified with the T161A mutation upon etorphine and with the S363A substitution upon DPDPE exposure. While both agonists induced a major receptor internalization, T161A and S363A impaired DOPr sequestration only for etorphine. However, similar level of S363 phosphorylation was measured between agonists. When CFP-tagged arrestin 3 was over-expressed, a similar profile of desensitization was measured for both agonists. In this context, all the 3 alanine mutations decreased etorphine-induced receptor desensitization. Using FRET, we showed similar interactions between WT hDOPr and arrestin 3 under DPDPE and etorphine stimulation which were delayed by both the Y318A and the S363A substitutions for etorphine. Finally, hDOPr recycling was qualitatively evaluated by microscopy and showed neither arrestin 3/hDOPr colocalization nor major impact of alanine mutations except for the S363A which impaired internalization and recycling for etorphine. The T161, Y318 and S363 residues of hDOPr could underlie the differential regulation promoted by DPDPE and etorphine.
Collapse
Affiliation(s)
- Elise Camacho
- Laboratoire de Signalisation, électrophysiologie et Imagerie des Lésions D'ischémie-reperfusion Myocardique, Normandie Univ, UNICAEN, Caen, France
| | - Nicolas Marie
- Centre National de Recherche Scientifique, Unité Mixte de Recherche 8206, Institut National de La Santé et de La Recherche Médicale. U705, Université Paris Descartes, Laboratoire de Neuropsychopharmacologie des Addictions, 4 Avenue de L'observatoire, 75006, Paris, France
| | - Quentin Dupas
- Laboratoire de Signalisation, électrophysiologie et Imagerie des Lésions D'ischémie-reperfusion Myocardique, Normandie Univ, UNICAEN, Caen, France
| | - Caroline Martel
- Laboratoire de Signalisation, électrophysiologie et Imagerie des Lésions D'ischémie-reperfusion Myocardique, Normandie Univ, UNICAEN, Caen, France
| | - Marie Nowoczyn
- Laboratoire de Signalisation, électrophysiologie et Imagerie des Lésions D'ischémie-reperfusion Myocardique, Normandie Univ, UNICAEN, Caen, France
| | - Nicolas Elie
- Plateau D'Histo-Imagerie Quantitative, CmaBio(3), SF 4206 ICORE, Normandie Univ, Caen, France
| | - Christophe Rochais
- Centre D'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ, UNICAEN, 14000, Caen, France
| | - Geza Töth
- Biological Research Centre, Institute of Biochemistry, Hungarian Academy of Sciences, Szeged, Hungary
| | - Stéphane Allouche
- Laboratoire de Signalisation, électrophysiologie et Imagerie des Lésions D'ischémie-reperfusion Myocardique, Normandie Univ, UNICAEN, Caen, France.
| |
Collapse
|
8
|
Benemei S, Lupi C, De Cesaris F, Lombardi N, Bettiol A, Chiarugi A, Geppetti P, Galli V, Pracucci C, Occupati B, Mannaioni G. Low-dose methadone for refractory chronic migraine accompanied by medication-overuse headache: a prospective cohort study. Neurol Sci 2020; 42:987-994. [PMID: 32691178 PMCID: PMC7870632 DOI: 10.1007/s10072-020-04602-3] [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: 02/28/2020] [Accepted: 07/14/2020] [Indexed: 12/04/2022]
Abstract
Objectives A refractory chronic migraine (RCM) accompanied by medication-overuse headache (MOH) is an extremely disabling disease. Evidence suggests that in selected patients, chronic opioids may be a valuable therapeutic option for RCM. The aim of the present study was to evaluate the effectiveness and safety of prophylaxis with low-dose methadone (LDM) in patients affected by RCM with continuous headache and MOH. Methods A prospective cohort study was performed between May 2012 and November 2015 at the Headache Center and Toxicology Unit of the Careggi University Hospital. Eligible patients were treated with prophylactic LDM and followed up for 12 months. Headache exacerbations, pain intensity, use of rescue medications, and occurrence of adverse drug reactions (ADRs) were recorded. Results Thirty patients (24 females, median age 48 years) were enrolled. Nineteen (63%) patients dropped out, mainly because of early ADRs (n = 10), including nausea, vomiting, and constipation. At last available follow-up, LDM was associated with a significant decrease in the number of headache attacks/month (from a median of 45 (interquartile range 30–150) to 16 (5–30), p < 0.001), in pain intensity (from 8.5 (8–9) to 5 (3–6), p < 0.001), and in the number of rescue medications consumed per month (from 95 (34–240) to 15 (3–28), p < 0.001). No misuse or diversion cases were observed. Conclusion LDM could represent a valuable and effective option in selected patients affected by RCM with continuous headache and MOH, although the frequency of early ADRs poses major safety concerns. Randomized controlled trials are needed to confirm the efficacy and safety of LDM prophylaxis.
Collapse
Affiliation(s)
- Silvia Benemei
- Headache Center, Careggi University Hospital, Florence, Italy.
| | - Chiara Lupi
- Headache Center, Careggi University Hospital, Florence, Italy
| | | | - Niccolò Lombardi
- NEUROFARBA Department, Toxicology Unit, Careggi University Hospital, University of Florence, Florence, Italy
| | - Alessandra Bettiol
- NEUROFARBA Department, Toxicology Unit, Careggi University Hospital, University of Florence, Florence, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, Headache Center, Careggi University Hospital, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Headache Center, Careggi University Hospital, University of Florence, Florence, Italy
| | - Valentina Galli
- Toxicology Unit, Careggi University Hospital, Florence, Italy
| | - Chiara Pracucci
- Toxicology Unit, Careggi University Hospital, Florence, Italy
| | | | - Guido Mannaioni
- NEUROFARBA Department, Toxicology Unit, Careggi University Hospital, University of Florence, Florence, Italy
| |
Collapse
|
9
|
Ma X, Chen R, Huang M, Wang W, Luo L, Kim DK, Jiang W, Xu T. DAMGO-induced μ opioid receptor internalization and recycling restore morphine sensitivity in tolerant rat. Eur J Pharmacol 2020; 878:173118. [PMID: 32320702 DOI: 10.1016/j.ejphar.2020.173118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 04/06/2020] [Accepted: 04/14/2020] [Indexed: 11/28/2022]
Abstract
This study investigated the effect of DAMGO-induced μ opioid receptor (MOR) internalization on morphine tolerance. Male Sprague-Dawley rats (200-250 g) aged 6-8 weeks were administered morphine via intrathecal (i.t.) injection (15 μg/10 μl twice daily for 6 days) to induce antinociceptive tolerance, which was evaluated using the tail-flick and paw-withdrawal tests. Response latency was calculated as the percentage of maximum possible effect (%MPE). A bolus of DAMGO was administered by i.t. injection on day 6, and the tail-flick and paw-withdrawal tests were carried out 24, 48, and 72 h later. Membrane and cytosolic MOR expression was assessed by western blotting. HEK293 cells were transfected with MOR-FLAG plasmid and after 6 days of morphine treatment (10 μM), the cells were treated with 1 μM DAMGO, and MOR localization was examined by immunofluorescence analysis 30 and 60 min later. Repeated morphine treatment induced tolerance after 5 days; however, i.t. DAMGO administration restored morphine sensitivity and enhanced acute morphine-induced antinociception after 24, 48, and 72 h. In HEK293 cells, DAMGO treatment stimulated MOR internalization after 30 min and MOR recycling to the membrane after 1 h. Membrane and cytoplasmic MOR expression in vivo was unchanged 24, 48, and 72 h after i.t. DAMGO injection. Morphine does not cause significant MOR internalization or downregulation, and can readily induce tolerance. DAMGO counters this effect by enhancing receptor endocytosis, thereby reversing morphine-induced antinociceptive tolerance and restoring its analgesic efficacy.
Collapse
Affiliation(s)
- Xiaqing Ma
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Rui Chen
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Min Huang
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Wenying Wang
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Limin Luo
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Dong Kwan Kim
- Department of Physiology, Konyang University College of Medicine, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Wei Jiang
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China.
| | - Tao Xu
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China; Department of Anesthesiology, Tongzhou People's Hospital, Nantong, 226300, China.
| |
Collapse
|
10
|
Ruland JG, Kirchhofer SB, Klindert S, Bailey CP, Bünemann M. Voltage modulates the effect of μ-receptor activation in a ligand-dependent manner. Br J Pharmacol 2020; 177:3489-3504. [PMID: 32297669 PMCID: PMC7348086 DOI: 10.1111/bph.15070] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/16/2020] [Accepted: 03/30/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Various GPCRs have been described as being modulated in a voltage-dependent manner. Opioid analgesics act via activation of μ receptors in various neurons. As neurons are exposed to large changes in membrane potential, we were interested in studying the effects of depolarization on μ receptor signalling. EXPERIMENTAL APPROACH We investigated potential voltage sensitivity of μ receptors in heterologous expression systems (HEK293T cells) using electrophysiology in combination with Förster resonance energy transfer-based assays. Depolarization-induced changes in signalling were also tested in physiological rat tissue containing locus coeruleus neurons. We applied depolarization steps across the physiological range of membrane potentials. KEY RESULTS Studying μ receptor function and signalling in cells, we discovered that morphine-induced signalling was strongly dependent on the membrane potential (VM ). This became apparent at the level of G-protein activation, G-protein coupled inwardly rectifying potassium channel (Kir 3.X) currents and binding of GPCR kinases and arrestin3 to μ receptors by a robust increase in signalling upon membrane depolarization. The pronounced voltage sensitivity of morphine-induced μ receptor activation was also observed at the level of Kir 3.X currents in rat locus coeruleus neurons. The efficacy of peptide ligands to activate μ receptors was not (Met-enkephalin) or only moderately ([D-Ala2 , N-Me-Phe4 , Gly5 -ol]-enkephalin) enhanced upon depolarization. In contrast, depolarization reduced the ability of the analgesic fentanyl to activate μ receptors. CONCLUSION AND IMPLICATIONS Our results indicate a strong ligand-dependent modulation of μ receptor activity by the membrane potential, suggesting preferential activity of morphine in neurons with high neuronal activity.
Collapse
Affiliation(s)
- Julia G Ruland
- Department of Pharmacology and Clinical Pharmacy, Philipps-University, Marburg, Germany
| | - Sina B Kirchhofer
- Department of Pharmacology and Clinical Pharmacy, Philipps-University, Marburg, Germany
| | - Sebastian Klindert
- Department of Pharmacology and Clinical Pharmacy, Philipps-University, Marburg, Germany.,Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Chris P Bailey
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Moritz Bünemann
- Department of Pharmacology and Clinical Pharmacy, Philipps-University, Marburg, Germany
| |
Collapse
|
11
|
Jullié D, Gondin AB, von Zastrow M, Canals M. Opioid Pharmacology under the Microscope. Mol Pharmacol 2020; 98:425-432. [PMID: 32198210 DOI: 10.1124/mol.119.119321] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/10/2020] [Indexed: 12/18/2022] Open
Abstract
The powerful analgesic effects of opioid drugs have captivated the interest of physicians and scientists for millennia, and the ability of opioid drugs to produce serious undesired effects has been recognized for a similar period of time (Kieffer and Evans, 2009). Many of these develop progressively with prolonged or repeated drug use and then persist, motivating particular interest in understanding how opioid drugs initiate adaptive or maladaptive modifications in neural function or regulation. Exciting advances have been made over the past several years in elucidating drug-induced changes at molecular, cellular, and physiologic scales of analysis. The present review will highlight some recent cellular studies that we believe bridge across scales and will focus on optical imaging approaches that put opioid drug action "under the microscope." SIGNIFICANCE STATEMENT: Opioid receptors are major pharmacological targets, but their signaling at the cellular level results from a complex interplay between pharmacology, regulation, subcellular localization, and membrane trafficking. This minireview discusses recent advances in understanding the cellular biology of opioid receptors, emphasizing particular topics discussed at the 50th anniversary of the International Narcotics Research Conference. Our goal is to highlight distinct signaling and regulatory properties emerging from the cellular biology of opioid receptors and discuss potential relevance to therapeutics.
Collapse
Affiliation(s)
- Damien Jullié
- Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California (D.J., M.v.Z.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia (A.B.G.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (M.C.); and Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom (M.C.)
| | - Arisbel B Gondin
- Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California (D.J., M.v.Z.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia (A.B.G.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (M.C.); and Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom (M.C.)
| | - Mark von Zastrow
- Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California (D.J., M.v.Z.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia (A.B.G.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (M.C.); and Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom (M.C.)
| | - Meritxell Canals
- Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California (D.J., M.v.Z.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia (A.B.G.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (M.C.); and Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom (M.C.)
| |
Collapse
|
12
|
Emery MA, Eitan S. Drug-specific differences in the ability of opioids to manage burn pain. Burns 2019; 46:503-513. [PMID: 31859093 DOI: 10.1016/j.burns.2019.03.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 03/04/2019] [Accepted: 03/30/2019] [Indexed: 12/17/2022]
Abstract
Burn injury pain is a significant public health problem. Burn injury treatment has improved tremendously in recent decades. However, an unintended consequence is that a larger number of patients now survive more severe injuries, and face intense pain that is very hard to treat. Although many efforts have been made to find alternative treatments, opioids remain the most effective medication available. Burn patients are frequently prescribed opioids in doses and durations that are significantly higher and longer than standard analgesic dosing guidelines. Despite this, many continue to experience unrelieved pain. They are also placed at a higher risk for developing dependence and opioid use disorder. Burn injury profoundly alters the functional state of the immune system. It also alters the expression levels of receptor, effector, and signaling molecules within the spinal cord's dorsal horn. These alterations could explain the reduced potency of opioids. However, recent studies demonstrate that different opioids signal preferentially via differential signaling pathways. This ligand-specific signaling by different opioids implies that burn injury may reduce the antinociceptive potency of opioids to different degrees, in a drug-specific manner. Indeed, recent findings hint at drug-specific differences in the ability of opioids to manage burn pain early after injury, as well as differences in their ability to prevent or treat the development of chronic and neuropathic pain. Here we review the current state of opioid treatment, as well as new findings that could potentially lead to opioid-based pain management strategies that may be significantly more effective than the current solutions.
Collapse
Affiliation(s)
- Michael A Emery
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA; Interdisciplinary Program in Neuroscience, Texas A&M Institute for Neuroscience (TAMIN), USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA; Interdisciplinary Program in Neuroscience, Texas A&M Institute for Neuroscience (TAMIN), USA.
| |
Collapse
|
13
|
Mortensen M, Krall J, Kongstad KT, Brygger BM, Lenzi O, Francotte P, Sørensen TE, Nielsen B, Jensen AA, Smart TG, Frølund B. Developing New 4-PIOL and 4-PHP Analogues for Photoinactivation of γ-Aminobutyric Acid Type A Receptors. ACS Chem Neurosci 2019; 10:4669-4684. [PMID: 31589403 DOI: 10.1021/acschemneuro.9b00478] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The critical roles played by GABAA receptors as inhibitory regulators of excitation in the central nervous system has been known for many years. Aberrant GABAA receptor function and trafficking deficits have also been associated with several diseases including anxiety, depression, epilepsy, and insomnia. As a consequence, important drug groups such as the benzodiazepines, barbiturates, and many general anesthetics have become established as modulators of GABAA receptor activity. Nevertheless, there is much we do not understand about the roles and mechanisms of GABAA receptors at neural network and systems levels. It is therefore crucial to develop novel technologies and especially chemical entities that can interrogate GABAA receptor function in the nervous system. Here, we describe the chemistry and characterization of a novel set of 4-PIOL and 4-PHP analogues synthesized with the aim of developing a toolkit of drugs that can photoinactivate GABAA receptors. Most of these new analogues show higher affinities/potencies compared with the respective lead compounds. This is indicative of cavernous areas being present near their binding sites that can be potentially associated with novel receptor interactions. The 4-PHP azide-analogue, 2d, possesses particularly impressive nanomolar affinity/potency and is an effective UV-inducible photoinhibitor of GABAA receptors with considerable potential for photocontrol of GABAA receptor function in situ.
Collapse
Affiliation(s)
- Martin Mortensen
- Department of Neuroscience, Physiology & Pharmacology , University College London , Gower Street , London WC1E 6BT , United Kingdom
| | - Jacob Krall
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , DK-2100 Copenhagen , Denmark
| | - Kenneth T Kongstad
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , DK-2100 Copenhagen , Denmark
| | - Benjamin M Brygger
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , DK-2100 Copenhagen , Denmark
| | - Ombretta Lenzi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , DK-2100 Copenhagen , Denmark
| | - Pierre Francotte
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , DK-2100 Copenhagen , Denmark.,Department of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM) , University of Liege , Avenue de l'Hôpital, 1, B36 , B-4000 Liège , Belgium
| | - Troels E Sørensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , DK-2100 Copenhagen , Denmark
| | - Birgitte Nielsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , DK-2100 Copenhagen , Denmark
| | - Anders A Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , DK-2100 Copenhagen , Denmark
| | - Trevor G Smart
- Department of Neuroscience, Physiology & Pharmacology , University College London , Gower Street , London WC1E 6BT , United Kingdom
| | - Bente Frølund
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , DK-2100 Copenhagen , Denmark
| |
Collapse
|
14
|
Arttamangkul S, Plazek A, Platt EJ, Jin H, Murray TF, Birdsong WT, Rice KC, Farrens DL, Williams JT. Visualizing endogenous opioid receptors in living neurons using ligand-directed chemistry. eLife 2019; 8:49319. [PMID: 31589142 PMCID: PMC6809603 DOI: 10.7554/elife.49319] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/06/2019] [Indexed: 12/11/2022] Open
Abstract
Identifying neurons that have functional opioid receptors is fundamental for the understanding of the cellular, synaptic and systems actions of opioids. Current techniques are limited to post hoc analyses of fixed tissues. Here we developed a fluorescent probe, naltrexamine-acylimidazole (NAI), to label opioid receptors based on a chemical approach termed ‘traceless affinity labeling’. In this approach, a high affinity antagonist naltrexamine is used as the guide molecule for a transferring reaction of acylimidazole at the receptor. This reaction generates a fluorescent dye covalently linked to the receptor while naltrexamine is liberated and leaves the binding site. The labeling induced by this reagent allowed visualization of opioid-sensitive neurons in rat and mouse brains without loss of function of the fluorescently labeled receptors. The ability to locate endogenous receptors in living tissues will aid considerably in establishing the distribution and physiological role of opioid receptors in the CNS of wild type animals.
Collapse
Affiliation(s)
- Seksiri Arttamangkul
- The Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Andrew Plazek
- Medicinal Chemistry Core, Oregon Health & Science University, Portland, United States
| | - Emily J Platt
- Department of Biochemistry and Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, United States
| | - Haihong Jin
- Medicinal Chemistry Core, Oregon Health & Science University, Portland, United States
| | - Thomas F Murray
- Department of Pharmacology, School of Medicine, Creighton University, Omaha, United States
| | - William T Birdsong
- The Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Kenner C Rice
- Drug Design and Synthesis Section, Intramural Research Program, NIDA and NIAAA, Bethesda, United States
| | - David L Farrens
- Department of Biochemistry and Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, United States
| | - John T Williams
- The Vollum Institute, Oregon Health & Science University, Portland, United States
| |
Collapse
|
15
|
Bellamy JR, Rubin BR, Zverovich A, Zhou Y, Contoreggi NH, Gray JD, McEwen BS, Kreek MJ, Milner TA. Sex and chronic stress differentially alter phosphorylated mu and delta opioid receptor levels in the rat hippocampus following oxycodone conditioned place preference. Neurosci Lett 2019; 713:134514. [PMID: 31560995 DOI: 10.1016/j.neulet.2019.134514] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 09/21/2019] [Indexed: 01/16/2023]
Abstract
Following oxycodone conditioned place preference (CPP) in naïve female and male Sprague Dawley rats, delta- and mu-opioid receptors (DORs and MORs) redistribute in hippocampal CA3 pyramidal cells and GABAergic interneurons in a manner that would promote opioid-associative learning processes, particularly in females. MORs and DORs similarly redistribute in CA3 and hilar neurons following chronic immobilization stress (CIS) in females, but not males, essentially "priming" the opioid system for oxycodone-associative learning. Following CIS, only females acquire oxycodone CPP. The present study determined whether sex and CIS differentially affect the levels of phosphorylated MORs and DORs (pMORs and pDORs) in the hippocampus following oxycodone CPP as phosphorylation is important for opioid receptor internationalization and trafficking. In naïve oxycodone-injected (Oxy) female rats, the density of pMOR-immunoreactivity (ir) was increased in CA1 stratum oriens and CA3a,b strata lucidum and radiatum compared to saline-injected (Sal)-females. Additionally, the density of pDOR-ir increased in the pyramidal cell layer and stratum radiatum of CA2/3a in Oxy-males compared to Sal-males. In CIS females that acquire CPP, pDOR-ir levels were increased in the CA2/3a. These findings indicate only rats that acquire oxycodone CPP have activated MORs and DORs in the hippocampus but that the subregion containing activated opioid receptors differs in females and males. These results are consistent with previously observed sex differences in the hippocampal opioid system following Oxy-CPP.
Collapse
Affiliation(s)
- Julia R Bellamy
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Batsheva R Rubin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Angelica Zverovich
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States.
| |
Collapse
|
16
|
DiCello JJ, Saito A, Rajasekhar P, Sebastian BW, McQuade RM, Gondin AB, Veldhuis NA, Canals M, Carbone SE, Poole DP. Agonist-dependent development of delta opioid receptor tolerance in the colon. Cell Mol Life Sci 2019; 76:3033-3050. [PMID: 30904952 PMCID: PMC11105391 DOI: 10.1007/s00018-019-03077-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/26/2019] [Accepted: 03/18/2019] [Indexed: 10/27/2022]
Abstract
The use of opioid analgesics is severely limited due to the development of intractable constipation, mediated through activation of mu opioid receptors (MOR) expressed by enteric neurons. The related delta opioid receptor (DOR) is an emerging therapeutic target for chronic pain, depression and anxiety. Whether DOR agonists also promote sustained inhibition of colonic transit is unknown. This study examined acute and chronic tolerance to SNC80 and ARM390, which were full and partial DOR agonists in neural pathways controlling colonic motility, respectively. Excitatory pathways developed acute and chronic tolerance to SNC80, whereas only chronic tolerance developed in inhibitory pathways. Both pathways remained functional after acute or chronic ARM390 exposure. Propagating colonic motor patterns were significantly reduced after acute or chronic SNC80 treatment, but not by ARM390 pre-treatment. These findings demonstrate that SNC80 has a prolonged inhibitory effect on propagating colonic motility. ARM390 had no effect on motor patterns and thus may have fewer gastrointestinal side-effects.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Benzamides/pharmacology
- Colon/drug effects
- Colon/physiology
- Drug Tolerance
- Electric Stimulation
- Mice
- Mice, Inbred C57BL
- Microscopy, Confocal
- Muscle Contraction/drug effects
- Neurons/metabolism
- Piperazines/pharmacology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Jesse J DiCello
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia.
| | - Ayame Saito
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Pradeep Rajasekhar
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Benjamin W Sebastian
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Rachel M McQuade
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Arisbel B Gondin
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Nicholas A Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Meritxell Canals
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Simona E Carbone
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Daniel P Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia.
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
17
|
Randesi M, Contoreggi NH, Zhou Y, Rubin BR, Bellamy JR, Yu F, Gray JD, McEwen BS, Milner TA, Kreek MJ. Sex Differences in Neuroplasticity- and Stress-Related Gene Expression and Protein Levels in the Rat Hippocampus Following Oxycodone Conditioned Place Preference. Neuroscience 2019; 410:274-292. [PMID: 31071414 DOI: 10.1016/j.neuroscience.2019.04.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/29/2019] [Accepted: 04/24/2019] [Indexed: 12/13/2022]
Abstract
Prescription opioid abuse is a serious public health issue. Recently, we showed that female and male Sprague-Dawley rats acquire conditioned place preference (CPP) to the mu opioid receptor agonist oxycodone. Anatomical analysis of the hippocampus from these rats unveiled sex differences in the opioid system in a way that would support excitation and opiate associative learning processes especially in females. In this study, we examined the expression and protein densities of opioid, plasticity, stress and related kinase and signaling molecules in the hippocampus of female and male rats following oxycodone CPP. Oxycodone CPP females have: a) increases in ARC (activity regulated cytoskeletal-associated protein)-immunoreactivity (ir) in CA3 pyramidal cells; b) decreases in Npy (neuropeptide Y) gene expression in the medial hippocampus but higher numbers of NPY-containing hilar interneurons compared to males; c) increases in Crhr2 (corticotropin releasing factor receptor 2) expression in CA2/3; d) increases in Akt1 (AKT serine/threonine kinase 1) expression in medial hippocampus; and e) decreases in phosphorylated MAPK (mitogen activated protein kinase)-ir in CA1 and dentate gyrus. Oxycodone CPP males have: a) increases in Bdnf (brain derived-neurotrophic factor) expression, which is known to be produced in granule cells, relative to females; b) elevated Mapk1 expression and pMAPK-ir in the dentate hilus which harbors newly generated granule cells; and c) increases in CRHR1-ir in CA3 pyramidal cell soma. These sex-specific changes in plasticity, stress and kinase markers in hippocampal circuitry parallel previously observed sex differences in the opioid system after oxycodone CPP.
Collapse
Affiliation(s)
- Matthew Randesi
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States of America
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States of America
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States of America
| | - Batsheva R Rubin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States of America
| | - Julia R Bellamy
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States of America
| | - Fangmin Yu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States of America
| | - Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States of America
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States of America
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States of America; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States of America.
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States of America
| |
Collapse
|
18
|
Eidson LN, Murphy AZ. Inflammatory mediators of opioid tolerance: Implications for dependency and addiction. Peptides 2019; 115:51-58. [PMID: 30890355 PMCID: PMC6863079 DOI: 10.1016/j.peptides.2019.01.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/24/2019] [Accepted: 01/30/2019] [Indexed: 12/27/2022]
Abstract
Each year, over 50 million Americans suffer from persistent pain, including debilitating headaches, joint pain, and severe back pain. Although morphine is amongst the most effective analgesics available for the management of severe pain, prolonged morphine treatment results in decreased analgesic efficacy (i.e., tolerance). Despite significant headway in the field, the mechanisms underlying the development of morphine tolerance are not well understood. The midbrain ventrolateral periaqueductal gray (vlPAG) is a primary neural substrate for the analgesic effects of morphine, as well as for the development of morphine tolerance. A growing body of literature indicates that activated glia (i.e., microglia and astrocytes) facilitate pain transmission and oppose morphine analgesia, making these cells important potential targets in the treatment of chronic pain. Morphine affects glia by binding to the innate immune receptor toll-like receptor 4 (TLR4), leading to the release of proinflammatory cytokines and opposition of morphine analgesia. Despite the established role of the vlPAG as an integral locus for the development of morphine tolerance, most studies have examined the role of glia activation within the spinal cord. Additionally, the role of TLR4 in the development of tolerance has not been elucidated. This review attempts to summarize what is known regarding the role of vlPAG glia and TLR4 in the development of morphine tolerance. These data, together, provide information about the mechanism by which central nervous system glia regulate morphine tolerance, and identify a potential therapeutic target for the enhancement of analgesic efficacy in the clinical treatment of chronic pain.
Collapse
Affiliation(s)
- Lori N Eidson
- Department of Physiology, Emory University, Atlanta, GA, 30322, United States
| | - Anne Z Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30308, United States.
| |
Collapse
|
19
|
Reich B, Zhou Y, Goldstein E, Srivats SS, Contoreggi NH, Kogan JF, McEwen BS, Kreek MJ, Milner TA, Gray JD. Chronic immobilization stress primes the hippocampal opioid system for oxycodone-associated learning in female but not male rats. Synapse 2019; 73:e22088. [PMID: 30632204 PMCID: PMC11548942 DOI: 10.1002/syn.22088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/27/2018] [Accepted: 01/06/2019] [Indexed: 12/21/2022]
Abstract
In adult female, but not male, Sprague Dawley rats, chronic immobilization stress (CIS) increases mossy fiber (MF) Leu-Enkephalin levels and redistributes delta- and mu-opioid receptors (DORs and MORs) in hippocampal CA3 pyramidal cells and GABAergic interneurons to promote excitation and learning processes following subsequent opioid exposure. Here, we demonstrate that CIS females, but not males, acquire conditioned place preference (CPP) to oxycodone and that CIS "primes" the hippocampal opioid system in females for oxycodone-associated learning. In CA3b, oxycodone-injected (Oxy) CIS females relative to saline-injected (Sal) CIS females exhibited an increase in the cytoplasmic and total densities of DORs in pyramidal cell dendrites so that they were similar to Sal- and Oxy-CIS males. Consistent with our earlier studies, Sal- and Oxy-CIS females but not CIS males had elevated DOR densities in MF-CA3 dendritic spines, which we have previously shown are important for opioid-mediated long-term potentiation. In the dentate gyrus, Oxy-CIS females had more DOR-labeled interneurons than Sal-CIS females. Moreover, Sal- and Oxy-CIS females compared to both groups of CIS males had elevated levels of DORs and MORs in GABAergic interneuron dendrites, suggesting capacity for greater synthesis or storage of these receptors in circuits important for opioid-mediated disinhibition. However, more plasmalemmal MORs were on large parvalbumin-containing dendrites of Oxy-CIS males compared to Sal-CIS males, suggesting a limited ability for increased granule cell disinhibition. These results suggest that low levels of DORs in MF-CA3 synapses and hilar GABAergic interneurons may contribute to the attenuation of oxycodone CPP in males exposed to CIS.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- CA3 Region, Hippocampal/cytology
- CA3 Region, Hippocampal/drug effects
- CA3 Region, Hippocampal/metabolism
- Conditioning, Classical
- Dendrites/metabolism
- Dentate Gyrus/cytology
- Dentate Gyrus/drug effects
- Dentate Gyrus/metabolism
- Female
- Interneurons/metabolism
- Male
- Oxycodone/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/metabolism
- Repetition Priming
- Restraint, Physical
- Stress, Psychological/metabolism
- Stress, Psychological/physiopathology
Collapse
Affiliation(s)
- Batsheva Reich
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Ellen Goldstein
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | - Sudarshan S. Srivats
- Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, P.O. Box 24144 - Doha, Qatar
| | - Natalina H. Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | - Joshua F. Kogan
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Bruce S. McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Jason D. Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| |
Collapse
|
20
|
Jeske NA. Dynamic Opioid Receptor Regulation in the Periphery. Mol Pharmacol 2019; 95:463-467. [PMID: 30723091 PMCID: PMC6442319 DOI: 10.1124/mol.118.114637] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/19/2018] [Indexed: 12/26/2022] Open
Abstract
Opioids serve a vital role in the current analgesic array of treatment options. They are useful in acute instances involving severe pain associated with trauma, surgery, and terminal diseases such as cancer. In the past three decades, multiple receptor isoforms and conformations have been reported throughout literature. Most of these studies conducted systemic analyses of opioid receptor function, often generalizing findings from receptor systems in central nervous tissue or exogenously expressing immortalized cell lines as common mechanisms throughout physiology. However, a culmination of innovative experimental data indicates that opioid receptor systems are differentially modulated depending on their anatomic expression profile. Importantly, opioid receptors expressed in the peripheral nervous system undergo regulation uncommon to similar receptors expressed in central nervous system tissues. This distinctive characteristic begs one to question whether peripheral opioid receptors maintain anatomically unique roles, and whether they may serve an analgesic advantage in providing pain relief without promoting addiction.
Collapse
Affiliation(s)
- Nathaniel A Jeske
- Departments of Oral and Maxillofacial Surgery, Pharmacology, and Physiology, Center for Biomedical Neuroscience, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
21
|
Ryan JD, Zhou Y, Contoreggi NH, Bshesh FK, Gray JD, Kogan JF, Ben KT, McEwen BS, Jeanne Kreek M, Milner TA. Sex Differences in the Rat Hippocampal Opioid System After Oxycodone Conditioned Place Preference. Neuroscience 2018; 393:236-257. [PMID: 30316908 PMCID: PMC6246823 DOI: 10.1016/j.neuroscience.2018.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/10/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022]
Abstract
Although opioid addiction has risen dramatically, the role of gender in addiction has been difficult to elucidate. We previously found sex-dependent differences in the hippocampal opioid system of Sprague-Dawley rats that may promote associative learning relevant to drug abuse. The present studies show that although female and male rats acquired conditioned place preference (CPP) to the mu-opioid receptor (MOR) agonist oxycodone (3 mg/kg, I.P.), hippocampal opioid circuits were differentially altered. In CA3, Leu-Enkephalin-containing mossy fibers had elevated levels in oxycodone CPP (Oxy) males comparable to those in females and sprouted in Oxy-females, suggesting different mechanisms for enhancing opioid sensitivity. Electron microscopy revealed that in Oxy-males delta opioid receptors (DORs) redistributed to mossy fiber-CA3 synapses in a manner resembling females that we previously showed is important for opioid-mediated long-term potentiation. Moreover, in Oxy-females DORs redistributed to CA3 pyramidal cell spines, suggesting the potential for enhanced plasticity processes. In Saline-injected (Sal) females, dentate hilar parvalbumin-containing basket interneuron dendrites had fewer MORs, however plasmalemmal and total MORs increased in Oxy-females. In dentate hilar GABAergic dendrites that contain neuropeptide Y, Sal-females compared to Sal-males had higher plasmalemmal DORs, and near-plasmalemmal DORs increased in Oxy-females. This redistribution of MORs and DORs within hilar interneurons in Oxy-females would potentially enhance disinhibition of granule cells via two different circuits. Together, these results indicate that oxycodone CPP induces sex-dependent redistributions of opioid receptors in hippocampal circuits in a manner facilitating opioid-associative learning processes and may help explain the increased susceptibility of females to opioid addiction acquisition and relapse.
Collapse
Affiliation(s)
- James D Ryan
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States.
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Farah K Bshesh
- Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, P.O. Box 24144 Doha, Qatar
| | - Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Joshua F Kogan
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Konrad T Ben
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States.
| |
Collapse
|
22
|
Llorca-Torralba M, Pilar-Cuéllar F, Bravo L, Bruzos-Cidon C, Torrecilla M, Mico JA, Ugedo L, Garro-Martínez E, Berrocoso E. Opioid Activity in the Locus Coeruleus Is Modulated by Chronic Neuropathic Pain. Mol Neurobiol 2018; 56:4135-4150. [PMID: 30284123 DOI: 10.1007/s12035-018-1361-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/20/2018] [Indexed: 12/16/2022]
Abstract
Pain affects both sensory and emotional aversive responses, often provoking depression and anxiety-related conditions when it becomes chronic. As the opioid receptors in the locus coeruleus (LC) have been implicated in pain, stress responses, and opioid drug effects, we explored the modifications to LC opioid neurotransmission in a chronic constriction injury (CCI) model of short- and long-term neuropathic pain (7 and 30 days after nerve injury). No significant changes were found after short-term CCI, yet after 30 days, CCI provoked an up-regulation of cAMP (cyclic 5'-adenosine monophosphate), pCREB (phosphorylated cAMP response element binding protein), protein kinase A, tyrosine hydroxylase, and electrical activity in the LC, as well as enhanced c-Fos expression. Acute mu opioid receptor desensitization was more intense in these animals, measured as the decline of the peak current caused by [Met5]-enkephalin and the reduction of forskolin-stimulated cAMP produced in response to DAMGO. Sustained morphine treatment did not markedly modify certain LC parameters in CCI-30d animals, such as [Met5]-enkephalin-induced potassium outward currents or burst activity and c-Fos rebound after naloxone precipitation, which may limit the development of some typical opioid drug-related adaptations. However, other phenomena were impaired by long-term CCI, including the reduction in forskolin-stimulated cAMP accumulation by DAMGO after naloxone precipitation in morphine dependent animals. Overall, this study suggests that long-term CCI leads to changes at the LC level that may contribute to the anxiodepressive phenotype that develops in these animals. Furthermore, opioid drugs produce complex adaptations in the LC in this model of chronic neuropathic pain.
Collapse
Affiliation(s)
- Meritxell Llorca-Torralba
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya, 21, 11009, Cádiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Fuencisla Pilar-Cuéllar
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), Departamento de Fisiología y Farmacología, Universidad de Cantabria, 39011, Santander, Spain
| | - Lidia Bravo
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya, 21, 11009, Cádiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Bruzos-Cidon
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940, Leioa, Spain
| | - María Torrecilla
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940, Leioa, Spain
| | - Juan A Mico
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya, 21, 11009, Cádiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Luisa Ugedo
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940, Leioa, Spain
| | - Emilio Garro-Martínez
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), Departamento de Fisiología y Farmacología, Universidad de Cantabria, 39011, Santander, Spain
| | - Esther Berrocoso
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya, 21, 11009, Cádiz, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.
- Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, University of Cádiz, 11510, Cádiz, Spain.
| |
Collapse
|
23
|
Medrano MC, Santamarta MT, Pablos P, Aira Z, Buesa I, Azkue JJ, Mendiguren A, Pineda J. Characterization of functional μ opioid receptor turnover in rat locus coeruleus: an electrophysiological and immunocytochemical study. Br J Pharmacol 2017; 174:2758-2772. [PMID: 28589556 DOI: 10.1111/bph.13901] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 05/16/2017] [Accepted: 05/26/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Regulation of μ receptor dynamics such as its trafficking is a possible mechanism underlying opioid tolerance that contributes to inefficient recycling of opioid responses. We aimed to characterize the functional turnover of μ receptors in the noradrenergic nucleus locus coeruleus (LC). EXPERIMENTAL APPROACH We measured opioid effect by single-unit extracellular recordings of LC neurons from rat brain slices. Immunocytochemical techniques were used to evaluate μ receptor trafficking. KEY RESULTS After near-complete, irreversible μ receptor inactivation with β-funaltrexamine (β-FNA), opioid effect spontaneously recovered in a rapid and efficacious manner. In contrast, α2 -adrenoceptor-mediated effect hardly recovered after receptor inactivation with the irreversible antagonist EEDQ. When the recovery of opioid effect was tested after various inactivating time schedules, we found that the longer the β-FNA pre-exposure, the less efficient and slower the functional μ receptor turnover became. Interestingly, μ receptor turnover was slower when β-FNA challenge was repeated in the same cell, indicating constitutive μ receptor recycling by trafficking from a depletable pool. Double immunocytochemistry confirmed the constitutive nature of μ receptor trafficking from a cytoplasmic compartment. The μ receptor turnover was slowed down when LC neuron calcium- or firing-dependent processes were prevented or vesicular protein trafficking was blocked by a low temperature or transport inhibitor. CONCLUSIONS AND IMPLICATIONS Constitutive trafficking of μ receptors from a depletable intracellular pool (endosome) may account for its rapid and efficient functional turnover in the LC. A finely-tuned regulation of μ receptor trafficking and endosomes could explain neuroadaptive plasticity to opioids in the LC.
Collapse
Affiliation(s)
- María Carmen Medrano
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - María Teresa Santamarta
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Patricia Pablos
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Zigor Aira
- Department of Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Itsaso Buesa
- Department of Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Jon Jatsu Azkue
- Department of Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Aitziber Mendiguren
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Joseba Pineda
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| |
Collapse
|
24
|
Abstract
Dendritic release of dopamine activates dopamine D2 autoreceptors, which are inhibitory G protein-coupled receptors (GPCRs), to decrease the excitability of dopamine neurons. This study used tagged D2 receptors to identify the localization and distribution of these receptors in living midbrain dopamine neurons. GFP-tagged D2 receptors were found to be unevenly clustered on the soma and dendrites of dopamine neurons within the substantia nigra pars compacta (SNc). Physiological signaling and desensitization of the tagged receptors were not different from wild type receptors. Unexpectedly, upon desensitization the tagged D2 receptors were not internalized. When tagged D2 receptors were expressed in locus coeruleus neurons, a desensitizing protocol induced significant internalization. Likewise, when tagged µ-opioid receptors were expressed in dopamine neurons they too were internalized. The distribution and lack of agonist-induced internalization of D2 receptors on dopamine neurons indicate a purposefully regulated localization of these receptors.
Collapse
|
25
|
Shiwarski DJ, Darr M, Telmer CA, Bruchez MP, Puthenveedu MA. PI3K class II α regulates δ-opioid receptor export from the trans-Golgi network. Mol Biol Cell 2017; 28:2202-2219. [PMID: 28566554 PMCID: PMC5531736 DOI: 10.1091/mbc.e17-01-0030] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/26/2017] [Accepted: 05/23/2017] [Indexed: 12/20/2022] Open
Abstract
The interplay between signaling and trafficking by G protein-coupled receptors (GPCRs) has focused mainly on endocytic trafficking. Whether and how surface delivery of newly synthesized GPCRs is regulated by extracellular signals is less understood. Here we define a signaling-regulated checkpoint at the trans-Golgi network (TGN) that controls the surface delivery of the delta opioid receptor (δR). In PC12 cells, inhibition of phosphoinositide-3 kinase (PI3K) activity blocked export of newly synthesized δR from the Golgi and delivery to the cell surface, similar to treatment with nerve growth factor (NGF). Depletion of class II phosphoinositide-3 kinase α (PI3K C2A), but not inhibition of class I PI3K, blocked δR export to comparable levels and attenuated δR-mediated cAMP inhibition. NGF treatment displaced PI3K C2A from the Golgi and optogenetic recruitment of the PI3K C2A kinase domain to the TGN-induced δR export downstream of NGF. Of importance, PI3K C2A expression promotes export of endogenous δR in primary trigeminal ganglion neurons. Taken together, our results identify PI3K C2A as being required and sufficient for δR export and surface delivery in neuronal cells and suggest that it could be a key modulator of a novel Golgi export checkpoint that coordinates GPCR delivery to the surface.
Collapse
Affiliation(s)
- Daniel J Shiwarski
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213.,Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Marlena Darr
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Cheryl A Telmer
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Marcel P Bruchez
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213.,Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213.,Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Manojkumar A Puthenveedu
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213 .,Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|
26
|
Porter-Stransky KA, Weinshenker D. Arresting the Development of Addiction: The Role of β-Arrestin 2 in Drug Abuse. J Pharmacol Exp Ther 2017; 361:341-348. [PMID: 28302862 DOI: 10.1124/jpet.117.240622] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/15/2017] [Indexed: 01/01/2023] Open
Abstract
The protein β-arrestin (βarr) 2 directly interacts with receptors and signaling pathways that mediate the behavioral effects of drugs of abuse, making it a prime candidate for therapeutic interventions. βarr2 drives desensitization and internalization of G protein-coupled receptors, including dopamine, opioid, and cannabinoid receptors, and it can also trigger G protein-independent intracellular signaling. βarr2 mediates several drug-induced behaviors, but the relationship is complex and dependent on the type of behavior (e.g., psychomotor versus reward), the class of drug (e.g., psychostimulant versus opioid), and the circuit being interrogated (e.g., brain region, cell type, and specific receptor ligand). Here we discuss the current state of research concerning the contribution of βarr2 to the psychomotor and rewarding effects of addictive drugs. Next we identify key knowledge gaps and suggest new tools and approaches needed to further elucidate the neuroanatomical substrates and neurobiological mechanisms to explain how βarr2 modulates behavioral responses to drugs of abuse, as well as its potential as a therapeutic target.
Collapse
Affiliation(s)
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
27
|
A PTEN-Regulated Checkpoint Controls Surface Delivery of δ Opioid Receptors. J Neurosci 2017; 37:3741-3752. [PMID: 28264976 DOI: 10.1523/jneurosci.2923-16.2017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/26/2017] [Accepted: 02/14/2017] [Indexed: 12/13/2022] Open
Abstract
The δ opioid receptor (δR) is a promising alternate target for pain management because δR agonists show decreased abuse potential compared with current opioid analgesics that target the μ opioid receptor. A critical limitation in developing δR as an analgesic target, however, is that δR agonists show relatively low efficacy in vivo, requiring the use of high doses that often cause adverse effects, such as convulsions. Here we tested whether intracellular retention of δR in sensory neurons contributes to this low δR agonist efficacy in vivo by limiting surface δR expression. Using direct visualization of δR trafficking and localization, we define a phosphatase and tensin homolog (PTEN)-regulated checkpoint that retains δR in the Golgi and decreases surface delivery in rat and mice sensory neurons. PTEN inhibition releases δR from this checkpoint and stimulates delivery of exogenous and endogenous δR to the neuronal surface both in vitro and in vivo PTEN inhibition in vivo increases the percentage of TG neurons expressing δR on the surface and allows efficient δR-mediated antihyperalgesia in mice. Together, we define a critical role for PTEN in regulating the surface delivery and bioavailability of the δR, explain the low efficacy of δR agonists in vivo, and provide evidence that active δR relocation is a viable strategy to increase δR antinociception.SIGNIFICANCE STATEMENT Opioid analgesics, such as morphine, which target the μ opioid receptor (μR), have been the mainstay of pain management, but their use is highly limited by adverse effects and their variable efficacy in chronic pain. Identifying alternate analgesic targets is therefore of great significance. Although the δ opioid receptor (δR) is an attractive option, a critical limiting factor in developing δR as a target has been the low efficacy of δR agonists. Why δR agonists show low efficacy is still under debate. This study provides mechanistic and functional data that intracellular localization of δR in neurons is a key factor that contributes to low agonist efficacy, and presents a proof of mechanism that relocating δR improves efficacy.
Collapse
|
28
|
Characterization of [ 3 H] oxymorphone binding sites in mouse brain: Quantitative autoradiography in opioid receptor knockout mice. Neurosci Lett 2017; 643:16-21. [DOI: 10.1016/j.neulet.2017.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/01/2017] [Accepted: 02/01/2017] [Indexed: 11/19/2022]
|
29
|
Gutwinski S, Schoofs N, Stuke H, Riemer TG, Wiers CE, Bermpohl F. Opioid tolerance in methadone maintenance treatment: comparison of methadone and levomethadone in long-term treatment. Harm Reduct J 2016; 13:7. [PMID: 26879120 PMCID: PMC4754801 DOI: 10.1186/s12954-016-0095-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/04/2016] [Indexed: 12/05/2022] Open
Abstract
Background This study aimed to investigate the development of opioid tolerance in patients receiving long-term methadone maintenance treatment (MMT). Methods A region-wide cross-sectional study was performed focusing on dosage and duration of treatment. Differences between racemic methadone and levomethadone were examined. All 20 psychiatric hospitals and all 110 outpatient clinics in Berlin licensed to offer MMT were approached in order to reach patients under MMT fulfilling the DSM IV criteria of opiate dependence. In the study, 720 patients treated with racemic methadone or levomethadone gave information on the dosage of treatment. Out of these, 679 patients indicated the duration of MMT. Results Treatment with racemic methadone was reported for 370 patients (54.5 %), with levomethadone for 309 patients (45.5 %). Mean duration of MMT was 7.5 years. We found a significant correlation between dosage and duration of treatment, both in a conjoint analysis for the two substances racemic methadone and levomethadone and for each substance separately. These effects remained significant when only patients receiving MMT for 1 year or longer were considered, indicating proceeding tolerance development in long-term treatment. When correlations were compared between racemic methadone and levomethadone, no significant difference was found. Conclusions Our data show a tolerance development under long-term treatment with both racemic methadone and levomethadone. Tolerance development did not differ significantly between the two substances.
Collapse
Affiliation(s)
- Stefan Gutwinski
- Department of Psychiatry and Psychotherapy, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Nikola Schoofs
- Department of Psychiatry and Psychotherapy, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Heiner Stuke
- Department of Psychiatry and Psychotherapy, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Thomas G Riemer
- Department of Psychiatry and Psychotherapy, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Corinde E Wiers
- Department of Psychiatry and Psychotherapy, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Felix Bermpohl
- Department of Psychiatry and Psychotherapy, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
30
|
Bowman SL, Soohoo AL, Shiwarski DJ, Schulz S, Pradhan AA, Puthenveedu MA. Cell-autonomous regulation of Mu-opioid receptor recycling by substance P. Cell Rep 2015; 10:1925-36. [PMID: 25801029 PMCID: PMC4494997 DOI: 10.1016/j.celrep.2015.02.045] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 10/31/2014] [Accepted: 02/18/2015] [Indexed: 10/26/2022] Open
Abstract
How neurons coordinate and reprogram multiple neurotransmitter signals is an area of broad interest. Here, we show that substance P (SP), a neuropeptide associated with inflammatory pain, reprograms opioid receptor recycling and signaling. SP, through activation of the neurokinin 1 (NK1R) receptor, increases the post-endocytic recycling of the mu-opioid receptor (MOR) in trigeminal ganglion (TG) neurons in an agonist-selective manner. SP-mediated protein kinase C (PKC) activation is both required and sufficient for increasing recycling of exogenous and endogenous MOR in TG neurons. The target of this cross-regulation is MOR itself, given that mutation of either of two PKC phosphorylation sites on MOR abolishes the SP-induced increase in recycling and resensitization. Furthermore, SP enhances the resensitization of fentanyl-induced, but not morphine-induced, antinociception in mice. Our results define a physiological pathway that cross-regulates opioid receptor recycling via direct modification of MOR and suggest a mode of homeostatic interaction between the pain and analgesic systems.
Collapse
|
31
|
Birdsong WT, Arttamangkul S, Bunzow JR, Williams JT. Agonist Binding and Desensitization of the μ-Opioid Receptor Is Modulated by Phosphorylation of the C-Terminal Tail Domain. Mol Pharmacol 2015; 88:816-24. [PMID: 25934731 DOI: 10.1124/mol.114.097527] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/30/2015] [Indexed: 11/22/2022] Open
Abstract
Sustained activation of G protein-coupled receptors can lead to a rapid decline in signaling through acute receptor desensitization. In the case of the μ-opioid receptor (MOPr), this desensitization may play a role in the development of analgesic tolerance. It is understood that phosphorylation of MOPr promotes association with β-arrestin proteins, which then facilitates desensitization and receptor internalization. Agonists that induce acute desensitization have been shown to induce a noncanonical high-affinity agonist binding state in MOPr, conferring a persistent memory of prior receptor activation. In the current study, live-cell confocal imaging was used to investigate the role of receptor phosphorylation in agonist binding to MOPr. A phosphorylation cluster in the C-terminal tail of MOPr was identified as a mediator of agonist-induced affinity changes in MOPr. This site is unique from the primary phosphorylation cluster responsible for β-arrestin binding and internalization. Electrophysiologic measurements of receptor function suggest that both phosphorylation clusters may play a parallel role during acute receptor desensitization. Desensitization was unaffected by alanine mutation of either phosphorylation cluster, but was largely eliminated when both clusters were mutated. Overall, this work suggests that there are multiple effects of MOPr phosphorylation that appear to regulate MOPr function: one affecting β-arrestin binding and a second affecting agonist binding.
Collapse
Affiliation(s)
| | | | - James R Bunzow
- Vollum Institute, Oregon Health & Science University, Portland, Oregon
| | - John T Williams
- Vollum Institute, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
32
|
Ceredig RA, Massotte D. Fluorescent knock-in mice to decipher the physiopathological role of G protein-coupled receptors. Front Pharmacol 2015; 5:289. [PMID: 25610398 PMCID: PMC4284998 DOI: 10.3389/fphar.2014.00289] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/12/2014] [Indexed: 12/21/2022] Open
Abstract
G protein-coupled receptors (GPCRs) modulate most physiological functions but are also critically involved in numerous pathological states. Approximately a third of marketed drugs target GPCRs, which places this family of receptors in the main arena of pharmacological pre-clinical and clinical research. The complexity of GPCR function demands comprehensive appraisal in native environment to collect in-depth knowledge of receptor physiopathological roles and assess the potential of therapeutic molecules. Identifying neurons expressing endogenous GPCRs is therefore essential to locate them within functional circuits whereas GPCR visualization with subcellular resolution is required to get insight into agonist-induced trafficking. Both remain frequently poorly investigated because direct visualization of endogenous receptors is often hampered by the lack of appropriate tools. Also, monitoring intracellular trafficking requires real-time visualization to gather in-depth knowledge. In this context, knock-in mice expressing a fluorescent protein or a fluorescent version of a GPCR under the control of the endogenous promoter not only help to decipher neuroanatomical circuits but also enable real-time monitoring with subcellular resolution thus providing invaluable information on their trafficking in response to a physiological or a pharmacological challenge. This review will present the animal models and discuss their contribution to the understanding of the physiopathological role of GPCRs. We will also address the drawbacks associated with this methodological approach and browse future directions.
Collapse
Affiliation(s)
- Rhian A Ceredig
- CNRS, Institut des Neurosciences Cellulaires et Intégratives, UPR 3212 Strasbourg, France
| | - Dominique Massotte
- CNRS, Institut des Neurosciences Cellulaires et Intégratives, UPR 3212 Strasbourg, France
| |
Collapse
|
33
|
Arttamangkul S, Birdsong W, Williams JT. Does PKC activation increase the homologous desensitization of μ opioid receptors? Br J Pharmacol 2015; 172:583-92. [PMID: 24697621 PMCID: PMC4292970 DOI: 10.1111/bph.12712] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 03/19/2014] [Accepted: 03/23/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE This study examined the role of agents known to activate PKC on morphine-induced desensitization of μ-opioid receptors (MOP receptors) in brain slices containing locus coeruleus neurons. EXPERIMENTAL APPROACH Intracellular recordings were obtained from rat locus coeruleus neurons. Two measurements were used to characterize desensitization, the decline in hyperpolarization induced by application of a saturating concentration of agonist (acute desensitization) and the decrease in hyperpolarization induced by a subsaturating concentration of [Met](5) enkephalin (ME) following washout of the saturating concentration (sustained desensitization). Internalization of MOP receptors was studied in brain slices prepared from transgenic mice expressing Flag-MOP receptors. The subcellular distribution of activated PKC was examined using a novel fluorescent sensor of PKC in HEK293 cells. KEY RESULTS The phorbol esters (PMA and PDBu) and muscarine increased acute desensitization induced by a saturating concentration of morphine and ME. These effects were not sensitive to staurosporine. Staurosporine did not block the decline in hyperpolarization induced by muscarine. PDBu and muscarine did not affect sustained desensitization induced by ME nor did phorbol esters or muscarine change the trafficking of MOP receptors induced by morphine or ME. The distribution of activated PKC measured in HEK293 cells differed depending on which phorbol ester was applied. CONCLUSIONS AND IMPLICATIONS This study demonstrates a distinct difference in two measurements that are often used to evaluate desensitization. The measure of decline correlated well with the reduction in peak amplitudes caused by PKC activators implicating the modification of other factors rather than MOP receptors. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
|
34
|
Allouche S, Noble F, Marie N. Opioid receptor desensitization: mechanisms and its link to tolerance. Front Pharmacol 2014; 5:280. [PMID: 25566076 PMCID: PMC4270172 DOI: 10.3389/fphar.2014.00280] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/02/2014] [Indexed: 02/04/2023] Open
Abstract
Opioid receptors (OR) are part of the class A of G-protein coupled receptors and the target of the opiates, the most powerful analgesic molecules used in clinic. During a protracted use, a tolerance to analgesic effect develops resulting in a reduction of the effectiveness. So understanding mechanisms of tolerance is a great challenge and may help to find new strategies to tackle this side effect. This review will summarize receptor-related mechanisms that could underlie tolerance especially receptor desensitization. We will focus on the latest data obtained on molecular mechanisms involved in opioid receptor desensitization: phosphorylation, receptor uncoupling, internalization, and post-endocytic fate of the receptor.
Collapse
Affiliation(s)
- Stéphane Allouche
- Laboratoire de Signalisation, Électrophysiologie et Imagerie des Lésions D'ischémie-Reperfusion Myocardique, Université de Caen, UPRES EA 4650, IFR 146 ICORE Caen, France
| | - Florence Noble
- Centre National de la Recherche Scientifique, ERL 3649 Paris, France ; Institut National de la Santé et de la Recherche Médicale, UMR-S 1124 Paris, France ; Université Paris Descartes, Neuroplasticité et Thérapies des Addictions Paris, France
| | - Nicolas Marie
- Centre National de la Recherche Scientifique, ERL 3649 Paris, France ; Institut National de la Santé et de la Recherche Médicale, UMR-S 1124 Paris, France ; Université Paris Descartes, Neuroplasticité et Thérapies des Addictions Paris, France
| |
Collapse
|
35
|
Duraffourd C, Kumala E, Anselmi L, Brecha NC, Sternini C. Opioid-induced mitogen-activated protein kinase signaling in rat enteric neurons following chronic morphine treatment. PLoS One 2014; 9:e110230. [PMID: 25302800 PMCID: PMC4193881 DOI: 10.1371/journal.pone.0110230] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 09/18/2014] [Indexed: 01/20/2023] Open
Abstract
Opioids, acting at μ opioid receptors, are commonly used for pain management. Chronic opioid treatment induces cellular adaptations, which trigger long-term side effects, including constipation mediated by enteric neurons. We tested the hypothesis that chronic opioid treatment induces alterations of μ opioid receptor signaling in enteric neurons, which are likely to serve as mechanisms underlying opioid-induced constipation. In cultured rat enteric neurons, either untreated (naïve) or exposed to morphine for 4 days (chronic), we compared the effect of morphine and DAMGO (D-Ala2,MePhe4,Gly-ol5 enkephalin) on μ opioid receptor internalization and downstream signaling by examining the activation of the mitogen-activated protein kinase/extracellular signal-regulated kinases 1 and 2 (MAPK/ERK) pathway, cAMP accumulation and transcription factor cAMP Response Element-Binding protein (CREB) expression. μ opioid receptor internalization and MAPK/ERK phosphorylation were induced by DAMGO, but not morphine in naïve neurons, and by both opioids in chronic neurons. MAPK/ERK activation was prevented by the receptor antagonist naloxone, by blocking receptor trafficking with hypertonic sucrose, dynamin inhibitor, or neuronal transfection with mutated dynamin, and by MAPK inhibitor. Morphine and DAMGO inhibited cAMP in naïve and chronic enteric neurons, and induced desensitization of cAMP signaling. Chronic morphine treatment suppressed desensitization of cAMP and MAPK signaling, increased CREB phosphorylation through a MAPK/ERK pathway and induced delays of gastrointestinal transit, which was prevented by MAPK/ERK blockade. This study showed that opioids induce endocytosis- and dynamin-dependent MAPK/ERK activation in enteric neurons and that chronic morphine treatment triggers changes at the receptor level and downstream signaling resulting in MAPK/ERK-dependent CREB activation. Blockade of this signaling pathway prevents the development of gastrointestinal motility impairment induced by chronic morphine treatment. These findings suggest that alterations in μ opioid receptor downstream signaling including MAPK/ERK pathway in enteric neurons chronically treated with morphine contribute to the development of opioid-induced constipation.
Collapse
Affiliation(s)
- Celine Duraffourd
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Erica Kumala
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Laura Anselmi
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Nicholas C. Brecha
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
| | - Catia Sternini
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
36
|
Santamarta MT, Llorente J, Mendiguren A, Pineda J. Involvement of neuronal nitric oxide synthase in desensitisation of µ-opioid receptors in the rat locus coeruleus. J Psychopharmacol 2014; 28:903-14. [PMID: 24961237 DOI: 10.1177/0269881114538542] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Nitric oxide (NO) has been recently shown to enhance µ-opioid receptor (MOR) desensitisation in locus coeruleus (LC) neurons. The aim of this study was to evaluate by single-unit extracellular recordings in rat brain slices whether the neuronal NO synthase is involved in MOR desensitisation in LC neurons. As expected, a high concentration of the opioid agonist Met(5)-enkephalin (ME; 10 µM, 10 min) strongly desensitised the inhibition induced by a test application of ME (0.8 µM, 1 min), whereas lower ME concentrations (1 and 3 µM) only weakly desensitised it. The neuronal NO synthase inhibitors 7-nitroindazole (10-100 µM), S-methyl-L-thiocitrulline (0.01-10 µM) and N(ω)-propyl-L-arginine (1-10 µM) attenuated ME (10 µM)-induced opioid desensitisation, although the endothelial NO synthase inhibitor N(5)-(1-iminoethyl)-L-ornithine (3-30 µM) failed to change it. The NO donor sodium nitroprusside (1 mM), but not its inactive analog potassium ferricyanide (1 mM), enhanced the ME (3 µM)-induced desensitisation and prevented the effect of S-methyl-L-thiocitrulline (10 µM). Sodium nitroprusside (1 mM) failed to change the desensitisation of α2-adrenoceptors by noradrenaline (100 µM, 10 min). These results suggest the contribution of NO and a neuronal type of NO synthase in homologous MOR desensitisation in rat LC neurons.
Collapse
Affiliation(s)
- María T Santamarta
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Javier Llorente
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Aitziber Mendiguren
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Joseba Pineda
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
37
|
Henderson G. The μ-opioid receptor: an electrophysiologist's perspective from the sharp end. Br J Pharmacol 2014; 172:260-7. [PMID: 24640948 DOI: 10.1111/bph.12633] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/02/2013] [Accepted: 12/10/2013] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Morphine, the prototypical opioid analgesic drug, produces its behavioural effects primarily through activation of μ-opioid receptors expressed in neurones of the central and peripheral nervous systems. This perspective provides a historical view of how, over the past 40 years, the use of electrophysiological recording techniques has helped to reveal the molecular mechanisms by which acute and chronic activation of μ-opioid receptors by morphine and other opioid drugs modify neuronal function. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- Graeme Henderson
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| |
Collapse
|
38
|
Cooke AE, Oldfield S, Krasel C, Mundell SJ, Henderson G, Kelly E. Morphine-induced internalization of the L83I mutant of the rat μ-opioid receptor. Br J Pharmacol 2014; 172:593-605. [PMID: 24697554 DOI: 10.1111/bph.12709] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 03/17/2014] [Accepted: 03/26/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Naturally occurring single-nucleotide polymorphisms (SNPs) within GPCRs can result in alterations in various pharmacological parameters. Understanding the regulation and function of endocytic trafficking of the μ-opioid receptor (MOP receptor) is of great importance given its implication in the development of opioid tolerance. This study has compared the agonist-dependent trafficking and signalling of L83I, the rat orthologue of a naturally occurring variant of the MOP receptor. EXPERIMENTAL APPROACH Cell surface elisa, confocal microscopy and immunoprecipitation assays were used to characterize the trafficking properties of the MOP-L83I variant in comparison with the wild-type receptor in HEK 293 cells. Functional assays were used to compare the ability of the L83I variant to signal to several downstream pathways. KEY RESULTS Morphine-induced internalization of the L83I MOP receptor was markedly increased in comparison with the wild-type receptor. The altered trafficking of this variant was found to be specific to morphine and was both G-protein receptor kinase- and dynamin-dependent. The enhanced internalization of L83I variant in response to morphine was not due to increased phosphorylation of serine 375, arrestin association or an increased ability to signal. CONCLUSIONS AND IMPLICATIONS These results suggest that morphine promotes a specific conformation of the L83I variant that makes it more liable to internalize in response to morphine, unlike the wild-type receptor that undergoes significantly less morphine-stimulated internalization, providing an example of a ligand-selective biased receptor. The presence of this SNP within an individual may consequently affect the development of tolerance and analgesic responses. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- A E Cooke
- School of Physiology and Pharmacology, University of Bristol, Bristol, BS8 1TD, UK
| | | | | | | | | | | |
Collapse
|
39
|
Massotte D. In vivo opioid receptor heteromerization: where do we stand? Br J Pharmacol 2014; 172:420-34. [PMID: 24666391 DOI: 10.1111/bph.12702] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 03/17/2014] [Accepted: 03/19/2014] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Opioid receptors are highly homologous GPCRs that modulate brain function at all levels of neural integration, including autonomous, sensory, emotional and cognitive processing. Opioid receptors functionally interact in vivo, but the underlying mechanisms involving direct receptor-receptor interactions, affecting signalling pathways or engaging different neuronal circuits, remain unsolved. Heteromer formation through direct physical interaction between two opioid receptors or between an opioid receptor and a non-opioid one has been postulated and can be characterized by specific ligand binding, receptor signalling and trafficking properties. However, despite numerous studies in heterologous systems, evidence for physical proximity in vivo is only available for a limited number of opioid heteromers, and their physiopathological implication remains largely unknown mostly due to the lack of appropriate tools. Nonetheless, data collected so far using endogenous receptors point to a crucial role for opioid heteromers as a molecular entity that could underlie human pathologies such as alcoholism, acute or chronic pain as well as psychiatric disorders. Opioid heteromers therefore stand as new therapeutic targets for the drug discovery field. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- D Massotte
- Institut des Neurosciences Cellulaires et Intégratives, INCI, Strasbourg, France
| |
Collapse
|
40
|
Connor M, Bagley EE, Chieng BC, Christie MJ. β-Arrestin-2 knockout prevents development of cellular μ-opioid receptor tolerance but does not affect opioid-withdrawal-related adaptations in single PAG neurons. Br J Pharmacol 2014; 172:492-500. [PMID: 24597632 DOI: 10.1111/bph.12673] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 02/24/2014] [Accepted: 03/01/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Tolerance to the behavioural effects of morphine is blunted in β-arrestin-2 knockout mice, but opioid withdrawal is largely unaffected. The cellular mechanisms of tolerance have been studied in some neurons from β-arrestin-2 knockouts, but tolerance and withdrawal mechanisms have not been examined at the cellular level in periaqueductal grey (PAG) neurons, which are crucial for central tolerance and withdrawal phenomena. EXPERIMENTAL APPROACH μ-Opioid receptor (MOPr) inhibition of voltage-gated calcium channel currents (ICa ) was examined by patch-clamp recordings from acutely dissociated PAG neurons from wild-type and β-arrestin-2 knockout mice treated chronically with morphine (CMT) or vehicle. Opioid withdrawal-induced activation of GABA transporter type 1 (GAT-1) currents was determined using perforated patch recordings from PAG neurons in brain slices. KEY RESULTS MOPr inhibition of ICa in PAG neurons was unaffected by β-arrestin-2 deletion. CMT impaired coupling of MOPrs to ICa in PAG neurons from wild-type mice, but this cellular tolerance was not observed in neurons from CMT β-arrestin-2 knockouts. However, β-arrestin-2 knockouts displayed similar opioid-withdrawal-induced activation of GAT-1 currents as wild-type PAG neurons. CONCLUSIONS AND IMPLICATIONS In β-arrestin-2 knockout mice, the central neurons involved in the anti-nociceptive actions of opioids also fail to develop cellular tolerance to opioids following chronic morphine. The results also provide the first cellular physiological evidence that opioid withdrawal is not disrupted by β-arrestin-2 deletion. However, the unaffected basal sensitivity to opioids in PAG neurons provides further evidence that changes in basal MOPr sensitivity cannot account for the enhanced acute nociceptive response to morphine reported in β-arrestin-2 knockouts. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- M Connor
- Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
41
|
Macey TA, Bobeck EN, Suchland KL, Morgan MM, Ingram SL. Change in functional selectivity of morphine with the development of antinociceptive tolerance. Br J Pharmacol 2014; 172:549-61. [PMID: 24666417 DOI: 10.1111/bph.12703] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 03/14/2014] [Accepted: 03/18/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Opioids, such as morphine, are the most effective treatment for pain but their efficacy is diminished with the development of tolerance following repeated administration. Recently, we found that morphine activated ERK in opioid-tolerant but not in naïve rats, suggesting that morphine activation of μ-opioid receptors is altered following repeated morphine administration. Here, we have tested the hypothesis that μ-opioid receptor activation of ERK in the ventrolateral periaqueductal gray (vlPAG) is dependent on dynamin, a protein implicated in receptor endocytosis. EXPERIMENTAL APPROACH Rats were made tolerant to repeated microinjections of morphine into the vlPAG. The effects of dynamin on ERK activation and antinociception were assessed by microinjecting myristoylated dominant-negative dynamin peptide (Dyn-DN) or a scrambled control peptide into the vlPAG. Microinjection of a fluorescent dermorphin analogue (DERM-A594) into the vlPAG was used to monitor μ-opioid receptor internalization. KEY RESULTS Morphine did not activate ERK and Dyn-DN administration had no effect on morphine-induced antinociception in saline-pretreated rats. In contrast, morphine-induced ERK activation in morphine-pretreated rats that was blocked by Dyn-DN administration. Dyn-DN also inhibited morphine antinociception. Finally, morphine reduced DERM-A594 internalization only in morphine-tolerant rats indicating that μ-opioid receptors were internalized and unavailable to bind DERM-A594. CONCLUSIONS AND IMPLICATIONS Repeated morphine administration increased μ-opioid receptor activation of ERK signalling via a dynamin-dependent mechanism. These results demonstrate that the balance of agonist signalling to G-protein and dynamin-dependent pathways is altered, effectively changing the functional selectivity of the agonist-receptor complex. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- T A Macey
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | | | | | | | | |
Collapse
|
42
|
Bourassa P, Tudashki HB, Pineyro G, Grandbois M, Gendron L. Label-free monitoring of μ-opioid receptor-mediated signaling. Mol Pharmacol 2014; 86:138-49. [PMID: 24874699 DOI: 10.1124/mol.114.093450] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
In this study, we used a combination of traditional signaling investigation approaches, bioluminescence resonance energy transfer (BRET) biosensors, and the label-free approach surface plasmon resonance (SPR) spectroscopy to monitor the signaling cascades of the μ-opioid receptor (MOP). In human embryonic kidney cells stably expressing a Flag-tagged version of human MOP, we compared the signals triggered by the noninternalizing and internalizing MOP agonists morphine and DAMGO (Tyr-D-Ala-Gly-N-methyl-Phe-Gly-ol), respectively. We studied three major and well described components of MOP signaling: receptor internalization, G protein coupling, and activation of extracellular signal-regulated kinase ERK1/ERK2. Our results show that morphine and DAMGO display different profiles of receptor internalization and a similar ability to trigger the phosphorylation of ERK1/ERK2. Our SPR analyses revealed that morphine and DAMGO evoke similar SPR signatures and that Gαi, cAMP-dependent pathways, and ERK1/ERK2 have key roles in morphine- and DAMGO-mediated signaling. Most interestingly, we found that the so-called MOP neutral antagonists CTOP (D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH(2)), naloxone, and naltrexone behave like partial agonists. Even more intriguing, BRET experiments indicate that CTAP (D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH(2)) induces similar conformational changes as naltrexone at the Gαi-βγ interface, whereas it appears as an inverse agonist based on its SPR response thus indicating distinct signaling mechanisms for the two ligands. Taken together, our results support the usefulness of label-free methods such as SPR to study whole-cell responses and signaling cascades triggered by G protein-coupled receptors and complement the conventional approaches by revealing cellular responses that would have been otherwise undetectable.
Collapse
Affiliation(s)
- Philippe Bourassa
- Départements de Physiologie et Biophysique (P.B., L.G.), Pharmacologie (H.B.T., M.G.), and Psychiatrie (G.P.), Centre de Recherche du CHU Ste-Justine (H.B.T, G.P.), Université de Montréal, Montreal, Quebec, Canada; Institut de Pharmacologie de Sherbrooke (M.G., L.G.), Centre de Recherche du CHU Sherbrooke (P.B., M.G., L.G.), Université de Sherbrooke (P.B., M.G., L.G.), Sherbrooke, Quebec, Canada; and Quebec Pain Research Network, Quebec City, Quebec, Canada (L.G.)
| | - Hanieh Bagheri Tudashki
- Départements de Physiologie et Biophysique (P.B., L.G.), Pharmacologie (H.B.T., M.G.), and Psychiatrie (G.P.), Centre de Recherche du CHU Ste-Justine (H.B.T, G.P.), Université de Montréal, Montreal, Quebec, Canada; Institut de Pharmacologie de Sherbrooke (M.G., L.G.), Centre de Recherche du CHU Sherbrooke (P.B., M.G., L.G.), Université de Sherbrooke (P.B., M.G., L.G.), Sherbrooke, Quebec, Canada; and Quebec Pain Research Network, Quebec City, Quebec, Canada (L.G.)
| | - Graciela Pineyro
- Départements de Physiologie et Biophysique (P.B., L.G.), Pharmacologie (H.B.T., M.G.), and Psychiatrie (G.P.), Centre de Recherche du CHU Ste-Justine (H.B.T, G.P.), Université de Montréal, Montreal, Quebec, Canada; Institut de Pharmacologie de Sherbrooke (M.G., L.G.), Centre de Recherche du CHU Sherbrooke (P.B., M.G., L.G.), Université de Sherbrooke (P.B., M.G., L.G.), Sherbrooke, Quebec, Canada; and Quebec Pain Research Network, Quebec City, Quebec, Canada (L.G.)
| | - Michel Grandbois
- Départements de Physiologie et Biophysique (P.B., L.G.), Pharmacologie (H.B.T., M.G.), and Psychiatrie (G.P.), Centre de Recherche du CHU Ste-Justine (H.B.T, G.P.), Université de Montréal, Montreal, Quebec, Canada; Institut de Pharmacologie de Sherbrooke (M.G., L.G.), Centre de Recherche du CHU Sherbrooke (P.B., M.G., L.G.), Université de Sherbrooke (P.B., M.G., L.G.), Sherbrooke, Quebec, Canada; and Quebec Pain Research Network, Quebec City, Quebec, Canada (L.G.)
| | - Louis Gendron
- Départements de Physiologie et Biophysique (P.B., L.G.), Pharmacologie (H.B.T., M.G.), and Psychiatrie (G.P.), Centre de Recherche du CHU Ste-Justine (H.B.T, G.P.), Université de Montréal, Montreal, Quebec, Canada; Institut de Pharmacologie de Sherbrooke (M.G., L.G.), Centre de Recherche du CHU Sherbrooke (P.B., M.G., L.G.), Université de Sherbrooke (P.B., M.G., L.G.), Sherbrooke, Quebec, Canada; and Quebec Pain Research Network, Quebec City, Quebec, Canada (L.G.)
| |
Collapse
|
43
|
Jaremko KM, Thompson NL, Reyes BAS, Jin J, Ebersole B, Jenney CB, Grigson PS, Levenson R, Berrettini WH, Van Bockstaele EJ. Morphine-induced trafficking of a mu-opioid receptor interacting protein in rat locus coeruleus neurons. Prog Neuropsychopharmacol Biol Psychiatry 2014; 50:53-65. [PMID: 24333843 PMCID: PMC3928604 DOI: 10.1016/j.pnpbp.2013.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 11/26/2013] [Accepted: 12/05/2013] [Indexed: 12/14/2022]
Abstract
Opiate addiction is a devastating health problem, with approximately 2million people currently addicted to heroin or non-medical prescription opiates in the United States alone. In neurons, adaptations in cell signaling cascades develop following opioid actions at the mu opioid receptor (MOR). A novel putative target for intervention involves interacting proteins that may regulate trafficking of MOR. Morphine has been shown to induce a re-distribution of a MOR-interacting protein Wntless (WLS, a transport molecule necessary for secretion of neurotrophic Wnt proteins), from cytoplasmic to membrane compartments in rat striatal neurons. Given its opiate-sensitivity and its well-characterized molecular and cellular adaptations to morphine exposure, we investigated the anatomical distribution of WLS and MOR in the rat locus coeruleus (LC)-norepinephrine (NE) system. Dual immunofluorescence microscopy was used to test the hypothesis that WLS is localized to noradrenergic neurons of the LC and that WLS and MOR co-exist in common LC somatodendritic processes, providing an anatomical substrate for their putative interactions. We also hypothesized that morphine would influence WLS distribution in the LC. Rats received saline, morphine or the opiate agonist [d-Ala2, N-Me-Phe4, Gly-ol5]-enkephalin (DAMGO), and tissue sections through the LC were processed for immunogold-silver detection of WLS and MOR. Statistical analysis showed a significant re-distribution of WLS to the plasma membrane following morphine treatment in addition to an increase in the proximity of gold-silver labels for MOR and WLS. Following DAMGO treatment, MOR and WLS were predominantly localized within the cytoplasmic compartment when compared to morphine and control. In a separate cohort of rats, brains were obtained from saline-treated or heroin self-administering male rats for pulldown co-immunoprecipitation studies. Results showed an increased association of WLS and MOR following heroin exposure. As the LC-NE system is important for cognition as well as decisions underlying substance abuse, adaptations in WLS trafficking and expression may play a role in modulating MOR function in the LC and contribute to the negative sequelae of opiate exposure on executive function.
Collapse
Affiliation(s)
- Kellie M Jaremko
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Nicholas L Thompson
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Beverly A S Reyes
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States.
| | - Jay Jin
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Brittany Ebersole
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Christopher B Jenney
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Patricia S Grigson
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Robert Levenson
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Wade H Berrettini
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, United States
| | - Elisabeth J Van Bockstaele
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| |
Collapse
|
44
|
Erbs E, Faget L, Scherrer G, Matifas A, Filliol D, Vonesch JL, Koch M, Kessler P, Hentsch D, Birling MC, Koutsourakis M, Vasseur L, Veinante P, Kieffer BL, Massotte D. A mu-delta opioid receptor brain atlas reveals neuronal co-occurrence in subcortical networks. Brain Struct Funct 2014; 220:677-702. [PMID: 24623156 PMCID: PMC4341027 DOI: 10.1007/s00429-014-0717-9] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 01/27/2014] [Indexed: 12/19/2022]
Abstract
Opioid receptors are G protein-coupled receptors (GPCRs) that modulate brain function at all levels of neural integration, including autonomic, sensory, emotional and cognitive processing. Mu (MOR) and delta (DOR) opioid receptors functionally interact in vivo, but whether interactions occur at circuitry, cellular or molecular levels remains unsolved. To challenge the hypothesis of MOR/DOR heteromerization in the brain, we generated redMOR/greenDOR double knock-in mice and report dual receptor mapping throughout the nervous system. Data are organized as an interactive database offering an opioid receptor atlas with concomitant MOR/DOR visualization at subcellular resolution, accessible online. We also provide co-immunoprecipitation-based evidence for receptor heteromerization in these mice. In the forebrain, MOR and DOR are mainly detected in separate neurons, suggesting system-level interactions in high-order processing. In contrast, neuronal co-localization is detected in subcortical networks essential for survival involved in eating and sexual behaviors or perception and response to aversive stimuli. In addition, potential MOR/DOR intracellular interactions within the nociceptive pathway offer novel therapeutic perspectives.
Collapse
Affiliation(s)
- Eric Erbs
- Department of Neurogenetics and Translational Medicine, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, BP10142, 67404 Illkirch cedex, France
| | - Lauren Faget
- Department of Neurogenetics and Translational Medicine, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, BP10142, 67404 Illkirch cedex, France
- Present Address: University of California, La Jolla, CA 92093 USA
| | - Gregory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Institute for Neuro-Innovation and Translational Neurosciences, Stanford University, Stanford, 94305 CA USA
| | - Audrey Matifas
- Department of Neurogenetics and Translational Medicine, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, BP10142, 67404 Illkirch cedex, France
| | - Dominique Filliol
- Department of Neurogenetics and Translational Medicine, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, BP10142, 67404 Illkirch cedex, France
| | - Jean-Luc Vonesch
- Imaging Centre, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, BP 10142, 1 rue Laurent Fries, 67404 Illkirch cedex, France
| | - Marc Koch
- Imaging Centre, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, BP 10142, 1 rue Laurent Fries, 67404 Illkirch cedex, France
| | - Pascal Kessler
- Imaging Centre, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, BP 10142, 1 rue Laurent Fries, 67404 Illkirch cedex, France
| | - Didier Hentsch
- Imaging Centre, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, BP 10142, 1 rue Laurent Fries, 67404 Illkirch cedex, France
| | | | - Manoussos Koutsourakis
- Institut Clinique de la Souris, 1 rue Laurent Fries, 67404 Illkirch cedex, France
- Present Address: Sanger Institute, Hinxton, Cambridge CB 10 1SA UK
| | - Laurent Vasseur
- Institut Clinique de la Souris, 1 rue Laurent Fries, 67404 Illkirch cedex, France
| | - Pierre Veinante
- Institut des Neurosciences Cellulaires et Intégratives CNRS UPR 3212, 5 rue Blaise Pascal, 67084 Strasbourg cedex 03, France
| | - Brigitte L. Kieffer
- Department of Neurogenetics and Translational Medicine, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, BP10142, 67404 Illkirch cedex, France
| | - Dominique Massotte
- Department of Neurogenetics and Translational Medicine, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, BP10142, 67404 Illkirch cedex, France
- Institut des Neurosciences Cellulaires et Intégratives CNRS UPR 3212, 5 rue Blaise Pascal, 67084 Strasbourg cedex 03, France
| |
Collapse
|
45
|
Tudashki HB, Robertson DN, Schiller PW, Pineyro G. Endocytic profiles of δ-opioid receptor ligands determine the duration of rapid but not sustained cAMP responses. Mol Pharmacol 2013; 85:148-61. [PMID: 24174495 DOI: 10.1124/mol.113.089003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Traditional assays that monitor cAMP inhibition by opioid receptor ligands require second-messenger accumulation over periods of 10-20 minutes. Since receptor regulation occurs within a similar time frame, such assays do not discriminate the actual signal from its modulation. Here we used bioluminescence resonance energy transfer to monitor inhibition of cAMP production by δ-opioid receptor (DOR) agonists in real time. cAMP inhibition elicited by different agonists over a period of 15 minutes was biphasic, with response buildup during the first 6 to 7 minutes, followed by a second phase of response decay or of no further increment. The rate at which the cAMP response disappeared was correlated with operational parameters describing ligand efficiency [log(τ/KA)] to promote Gαi activation, as well as with ligand ability to promote internalization during the time course of the assay. Thus, ligands that displayed low signaling efficiency and poor sequestration(SB235863 ([8R-(4bS*,8aα,8aβ,12bβ)]7,10-dimethyl-1-methoxy-11-(2-ethylpropyl)oxycarbonyl 5,6,7,8,12,12b-hexahydro-(9H)-4,8-methanobenzofuro[3,2-e]pyrrolo[2,3-g]isoquinoline hydrochloride), morphine) had minimal or no response decay. On the other hand, the decay rate was pronounced for deltorphin II, [d-Pen(2), d-pen(5)]-enkephalin, met-enkephalin, and SNC-80 ((+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide), which displayed high signaling efficiency and internalization. Moreover, inhibition of internalization by dynasore reduced or abolished response decay by internalizing ligands. Unlike acute responses, endocytic profiles were not predictive of whether an agonist would induce prolonged cAMP inhibition over sustained (30-120 minutes) DOR stimulation. Taken together, the data indicate that ligand ability to evoke G-protein activation or promote endocytosis was predictive of response duration over short, but not over sustained periods of cAMP inhibition.
Collapse
Affiliation(s)
- Hanieh Bagheri Tudashki
- Centre de Recherche, Sainte-Justine Hospital, Montréal, Québec, Canada (H.B.T., D.N.R., G.P.); Department of Pharmacology, Faculty of Medicine, University of Montréal, Montréal, Québec, Canada (H.B.T., P.W.S., G.P.); and Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montréal, Québec, Canada (P.W.S.)
| | | | | | | |
Collapse
|
46
|
Barwatt JW, Hofford RS, Emery MA, Bates MLS, Wellman PJ, Eitan S. Differential effects of methadone and buprenorphine on the response of D2/D3 dopamine receptors in adolescent mice. Drug Alcohol Depend 2013; 132:420-6. [PMID: 23932842 DOI: 10.1016/j.drugalcdep.2013.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 06/19/2013] [Accepted: 07/12/2013] [Indexed: 11/18/2022]
Abstract
BACKGROUND There is a lack of studies that examine the effects of opioid maintenance drugs on the developing adolescent brain, limiting the ability of physicians to conduct a science-based risk assessment on the appropriateness of these treatments for that age group. Our recent observations indicate higher potential risks in repeated exposure to morphine during adolescence, specifically to the D2/D3 dopamine receptors' signaling. Disturbances in dopaminergic signaling could have broader implications for long-term mental health. Thus, this study examined whether buprenorphine and methadone differentially alter the responses of the D2/D3 dopamine receptors in adolescents. METHODS Adolescent mice were orally administered buprenorphine (0.1-0.4 mg/kg), methadone (25-100 mg/kg), or saline once daily for 6 days. Two hours or three days later, the mice were tested for their locomotor response to 10 mg/kg quinpirole, a D2/D3 dopamine receptor agonist. RESULTS Buprenorphine-treated adolescent mice did not significantly differ from control drug-naïve animals in their response to quinpirole. However, an enhanced response was observed in methadone-treated adolescent animals. This enhanced locomotion was significantly higher two hours following the final dose of methadone, as compared to three days afterwards. CONCLUSIONS This study suggests that exposure to various opioids carries differential probabilities of altering the highly sensitive neurochemistry of adolescent brains. Methadone exposure disturbs the D2-like receptor's response, indicating a potential risk in administering methadone to adolescents (either for the treatment of opioid dependency/abuse or for pain management). In contrast, buprenorphine appears to have a significantly lower effect on the behavioral sensitivity of D2/D3 dopamine receptors in adolescents.
Collapse
Affiliation(s)
- J William Barwatt
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | | | | | | | | | | |
Collapse
|
47
|
Gantz SC, Bunzow JR, Williams JT. Spontaneous inhibitory synaptic currents mediated by a G protein-coupled receptor. Neuron 2013; 78:807-12. [PMID: 23764286 DOI: 10.1016/j.neuron.2013.04.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2013] [Indexed: 11/30/2022]
Abstract
G protein-coupled receptors (GPCRs) affect many physiological processes by modulating both intrinsic membrane conductances and synaptic transmission. This study describes spontaneous miniature inhibitory postsynaptic currents mediated by vesicular dopamine release acting locally on metabotropic D2 receptors leading to the activation of a G protein-coupled inwardly rectifying potassium conductance. Thus, individual exocytotic events result in spontaneous GPCR-mediated transmission, similar to synaptic activation of classical ligand-gated ion channels.
Collapse
Affiliation(s)
- Stephanie C Gantz
- Vollum Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | | | | |
Collapse
|
48
|
Niikura K, Ho A, Kreek MJ, Zhang Y. Oxycodone-induced conditioned place preference and sensitization of locomotor activity in adolescent and adult mice. Pharmacol Biochem Behav 2013; 110:112-6. [PMID: 23827650 DOI: 10.1016/j.pbb.2013.06.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/25/2013] [Accepted: 06/22/2013] [Indexed: 10/26/2022]
Abstract
Nonmedical use of the prescription opioid oxycodone has become a major public health problem in the United States, with special concern for adolescents. Although adults and adolescents have different sensitivities for drugs, little is known about the rewarding effects of oxycodone in adolescents compared to adults, even in rodent models. Here, we investigate sensitivity to oxycodone by the conditioned place preference assay of conditioned reward, and effect on the locomotor activity in adolescent (4 weeks old) and adult (10 weeks old) C57BL/6J mice. Mice of both ages were trained with multiple doses of oxycodone (0, 0.3, 1, and 3 mg/kg) and showed conditioned preference in a dose-dependent manner. The adult mice developed conditioned preference to the lowest dose tested (0.3 mg/kg), but adolescent mice did not. Dose-dependent oxycodone-induced increases in locomotor activity were observed across the conditioning session. Interestingly, adolescent mice developed greater sensitization to the locomotor-activating effects of oxycodone than adult mice. Thus differences in sensitivity to oxycodone, such as the lower initial sensitivity for conditioned preference but greater locomotor sensitization in adolescent mice, may indicate contributing factors in oxycodone abuse and later addiction in human adolescents.
Collapse
Affiliation(s)
- Keiichi Niikura
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| | | | | | | |
Collapse
|
49
|
Beerepoot P, Lam VM, Salahpour A. Measurement of G protein-coupled receptor surface expression. J Recept Signal Transduct Res 2013; 33:162-5. [DOI: 10.3109/10799893.2013.781625] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
50
|
Anselmi L, Jaramillo I, Palacios M, Huynh J, Sternini C. Ligand-induced μ opioid receptor internalization in enteric neurons following chronic treatment with the opiate fentanyl. J Neurosci Res 2013; 91:854-60. [PMID: 23553842 DOI: 10.1002/jnr.23214] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 01/11/2013] [Accepted: 01/14/2013] [Indexed: 12/19/2022]
Abstract
Morphine differs from most opiates its poor ability to internalize μ opioid receptors (μORs). However, chronic treatment with morphine produces adaptational changes at the dynamin level, which enhance the efficiency of acute morphine stimulation to promote μOR internalization in enteric neurons. This study tested the effect of chronic treatment with fentanyl, a μOR-internalizing agonist, on ligand-induced endocytosis and the expression of the intracellular trafficking proteins, dynamin and β-arrestin, in enteric neurons using organotypic cultures of the guinea pig ileum. In enteric neurons from guinea pigs chronically treated with fentanyl, μOR immunoreactivity was predominantly at the cell surface after acute exposure to morphine with a low level of μOR translocation, slightly higher than in neurons from naïve animals. This internalization was not due to morphine's direct effect, because it was also observed in neurons exposed to medium alone. By contrast, D-Ala2-N-Me-Phe4-Gly-ol5-enkephalin (DAMGO), a potent μOR-internalizing agonist, induced pronounced and rapid μOR endocytosis in enteric neurons from animals chronically treated with fentanyl or from naïve animals. Chronic fentanyl treatment did not alter dynamin or β-arrestin expression. These findings indicate that prolonged activation of μORs with an internalizing agonist such as fentanyl does not enhance the ability of acute morphine to trigger μOR endocytosis or induce changes in intracellular trafficking proteins, as observed with prolonged activation of μORs with a poorly internalizing agonist such as morphine. Cellular adaptations induced by chronic opiate treatment might be ligand dependent and vary with the agonist efficiency to induce receptor internalization.
Collapse
Affiliation(s)
- Laura Anselmi
- CURE Digestive Diseases Research Center, Digestive Diseases Division, Los Angeles, CA 90073, USA
| | | | | | | | | |
Collapse
|