1
|
Underwood O, Fritzwanker S, Glenn J, Blum NK, Batista-Gondin A, Drube J, Hoffmann C, Briddon SJ, Schulz S, Canals M. Key phosphorylation sites for robust β-arrestin2 binding at the MOR revisited. Commun Biol 2024; 7:933. [PMID: 39095612 PMCID: PMC11297201 DOI: 10.1038/s42003-024-06571-1] [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: 01/09/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024] Open
Abstract
Desensitisation of the mu-opioid receptor (MOR) is proposed to underlie the initiation of opioid analgesic tolerance and previous work has shown that agonist-induced phosphorylation of the MOR C-tail contributes to this desensitisation. Moreover, phosphorylation is important for β-arrestin recruitment to the receptor, and ligands of different efficacies induce distinct phosphorylation barcodes. The C-tail 370TREHPSTANT379 motif harbours Ser/Thr residues important for these regulatory functions. 375Ser is the primary phosphorylation site of a ligand-dependent, hierarchical, and sequential process, whereby flanking 370Thr, 376Thr and 379Thr get subsequently and rapidly phosphorylated. Here we used GRK KO cells, phosphosite specific antibodies and site-directed mutagenesis to evaluate the contribution of the different GRK subfamilies to ligand-induced phosphorylation barcodes and β-arrestin2 recruitment. We show that both GRK2/3 and GRK5/6 subfamilies promote phosphorylation of 370Thr and 375Ser. Importantly, only GRK2/3 induce phosphorylation of 376Thr and 379Thr, and we identify these residues as key sites to promote robust β-arrestin recruitment to the MOR. These data provide insight into the mechanisms of MOR regulation and suggest that the cellular complement of GRK subfamilies plays an important role in determining the tissue responses of opioid agonists.
Collapse
Affiliation(s)
- Owen Underwood
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, Midlands, UK
| | - Sebastian Fritzwanker
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Jaqueline Glenn
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, Midlands, UK
| | - Nina Kathleen Blum
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Arisbel Batista-Gondin
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Victoria, Australia
| | - Julia Drube
- Institut fur Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Carsten Hoffmann
- Institut fur Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Stephen J Briddon
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, Midlands, UK
| | - Stefan Schulz
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
- 7TM Antibodies GmbH, Hans-Knöll-Straße 6, D-07745, Jena, Germany
| | - Meritxell Canals
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, Midlands, UK.
| |
Collapse
|
2
|
Avila-Zozaya M, Zachariou V. Genetic mouse models in opioid research: current status and future directions. J Neural Transm (Vienna) 2024; 131:491-494. [PMID: 38436758 DOI: 10.1007/s00702-024-02762-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
Synthetic and semi-synthetic opioids are prescribed for the management of severe pain conditions, but their long-term use is often leading to physical dependence and addiction disorders. Understanding the complex neurobiology of the opioid system in preclinical models will be essential for the development of safe and efficacious analgesics. With rising numbers of synthetic opioid users and overdose cases, a better understanding of the neuroanatomical and cellular pathways associated with physical dependence and addiction is expected to guide treatment approaches for opioid use disorders. In this commentary, we highlight the importance of advanced genetic mouse models for studying the regional effects of opioid receptors, and we discuss the need of genetic mouse models for the investigation of the regional, circuit and cell compartment-specific role of intracellular mediators of opioid actions.
Collapse
Affiliation(s)
- Monserrat Avila-Zozaya
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Venetia Zachariou
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
3
|
Kise R, Inoue A. GPCR signaling bias: an emerging framework for opioid drug development. J Biochem 2024; 175:367-376. [PMID: 38308136 DOI: 10.1093/jb/mvae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/18/2024] [Accepted: 01/31/2024] [Indexed: 02/04/2024] Open
Abstract
Biased signaling, also known as functional selectivity, has emerged as an important concept in drug development targeting G-protein-coupled receptors (GPCRs). Drugs that provoke biased signaling are expected to offer an opportunity for enhanced therapeutic effectiveness with minimized side effects. Opioid analgesics, whilst exerting potent pain-relieving effects, have become a social problem owing to their serious side effects. For the development of safer pain medications, there has been extensive exploration of agonists with a distinct balance of G-protein and β-arrestin (βarr) signaling. Recently, several approaches based on protein-protein interactions have been developed to precisely evaluate individual signal pathways, paving the way for the comprehensive analysis of biased signals. In this review, we describe an overview of bias signaling in opioid receptors, especially the μ-opioid receptor (MOR), and how to evaluate signaling bias in the GPCR field. We also discuss future directions for rational drug development through the integration of diverse signal datasets.
Collapse
Affiliation(s)
- Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
4
|
Chvilicek MM, Seguin A, Lathen DR, Titos I, Cummins‐Beebee PN, Pabon MA, Miščević M, Nickel E, Merrill CB, Rodan AR, Rothenfluh A. Large analysis of genetic manipulations reveals an inverse correlation between initial alcohol resistance and rapid tolerance phenotypes. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12884. [PMID: 38968320 PMCID: PMC10825885 DOI: 10.1111/gbb.12884] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 07/07/2024]
Abstract
Tolerance occurs when, following an initial experience with a substance, more of the substance is required subsequently to induce identical behavioral effects. Tolerance is not well-understood, and numerous researchers have turned to model organisms, particularly Drosophila melanogaster, to unravel its mechanisms. Flies have high translational relevance for human alcohol responses, and there is substantial overlap in disease-causing genes between flies and humans, including those associated with Alcohol Use Disorder. Numerous Drosophila tolerance mutants have been described; however, approaches used to identify and characterize these mutants have varied across time and labs and have mostly disregarded any impact of initial resistance/sensitivity to ethanol on subsequent tolerance development. Here, we analyzed our own, as well as data published by other labs to uncover an inverse correlation between initial ethanol resistance and tolerance phenotypes. This inverse correlation suggests that initial resistance phenotypes can explain many 'perceived' tolerance phenotypes, thus classifying such mutants as 'secondary' tolerance mutants. Additionally, we show that tolerance should be measured as a relative increase in time to sedation between an initial and second exposure rather than an absolute change in time to sedation. Finally, based on our analysis, we provide a method for using a linear regression equation to assess the residuals of potential tolerance mutants. These residuals provide predictive insight into the likelihood of a mutant being a 'primary' tolerance mutant, where a tolerance phenotype is not solely a consequence of initial resistance, and we offer a framework for understanding the relationship between initial resistance and tolerance.
Collapse
Affiliation(s)
- Maggie M. Chvilicek
- Department of Psychiatry, Huntsman Mental Health Institute, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Neuroscience Graduate ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - Alexandra Seguin
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Daniel R. Lathen
- Department of Psychiatry, Huntsman Mental Health Institute, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Neuroscience Graduate ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - Iris Titos
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Pearl N. Cummins‐Beebee
- Department of Psychiatry, Huntsman Mental Health Institute, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Neuroscience Graduate ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - Miguel A. Pabon
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Maša Miščević
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Present address:
Department of Neuroscience, Physiological Sciences Graduate Interdisciplinary ProgramUniversity of ArizonaTucsonArizonaUSA
| | - Emily Nickel
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Collin B. Merrill
- Department of Psychiatry, Huntsman Mental Health Institute, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Aylin R. Rodan
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Division of Nephrology, Department of Internal Medicine, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Medical ServiceVeterans Affairs Salt Lake City Health Care SystemSalt Lake CityUtahUSA
- Department of Human Genetics, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Adrian Rothenfluh
- Department of Psychiatry, Huntsman Mental Health Institute, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Neuroscience Graduate ProgramUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Department of Human Genetics, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Department of Neurobiology, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
5
|
Chvilicek MM, Seguin A, Lathen DR, Titos I, Cummins-Beebe PN, Pabon MA, Miscevic M, Nickel EA, Merrill CB, Rodan AR, Rothenfluh A. Large genetic analysis of alcohol resistance and tolerance reveals an inverse correlation and suggests 'true' tolerance mutants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561599. [PMID: 37873285 PMCID: PMC10592763 DOI: 10.1101/2023.10.09.561599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Tolerance occurs when, following an initial experience with a substance, more of the substance is required subsequently to induce the same behavioral effects. Tolerance is historically not well-understood, and numerous researchers have turned to model organisms, particularly Drosophila melanogaster, to unravel its mechanisms. Flies have high translational relevance for human alcohol responses, and there is substantial overlap in disease-causing genes between flies and humans, including those associated with Alcohol Use Disorder. Numerous Drosophila tolerance mutants have been described; however, approaches used to identify and characterize these mutants have varied across time and between labs and have mostly disregarded any impact of initial resistance/sensitivity to ethanol on subsequent tolerance development. Here, we have analyzed a large amount of data - our own published and unpublished data and data published by other labs - to uncover an inverse correlation between initial ethanol resistance and tolerance phenotypes. This inverse correlation suggests that initial resistance phenotypes can explain many 'perceived' tolerance phenotypes. Additionally, we show that tolerance should be measured as a relative increase in time to sedation between an initial and second exposure rather than an absolute change in time to sedation. Finally, based on our analysis, we provide a method for using a linear regression equation to assess the residuals of potential tolerance mutants. We show that these residuals provide predictive insight into the likelihood of a mutant being a 'true' tolerance mutant, and we offer a framework for understanding the relationship between initial resistance and tolerance.
Collapse
Affiliation(s)
- Maggie M. Chvilicek
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, USA
| | - Alexandra Seguin
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
| | - Daniel R. Lathen
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, USA
| | - Iris Titos
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
| | - Pearl N Cummins-Beebe
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, USA
| | - Miguel A. Pabon
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
| | - Masa Miscevic
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
| | - Emily A. Nickel
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
| | - Collin B Merrill
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
| | - Aylin R. Rodan
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
- Division of Nephrology, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, USA
- Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, USA
- Department of Human Genetics, School of Medicine, University of Utah, Salt Lake City, USA
| | - Adrian Rothenfluh
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, USA
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
- Department of Human Genetics, School of Medicine, University of Utah, Salt Lake City, USA
- Department of Neurobiology, School of Medicine, University of Utah, Salt Lake City, USA
| |
Collapse
|
6
|
Coutens B, Ingram SL. Key differences in regulation of opioid receptors localized to presynaptic terminals compared to somas: Relevance for novel therapeutics. Neuropharmacology 2023; 226:109408. [PMID: 36584882 PMCID: PMC9898207 DOI: 10.1016/j.neuropharm.2022.109408] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/05/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022]
Abstract
Opioid receptors are G protein-coupled receptors (GPCRs) that regulate activity within peripheral, subcortical and cortical circuits involved in pain, reward, and aversion processing. Opioid receptors are expressed in both presynaptic terminals where they inhibit neurotransmitter release and postsynaptic locations where they act to hyperpolarize neurons and reduce activity. Agonist activation of postsynaptic receptors at the plasma membrane signal via ion channels or cytoplasmic second messengers. Agonist binding initiates regulatory processes that include phosphorylation by G protein receptor kinases (GRKs) and recruitment of beta-arrestins that desensitize and internalize the receptors. Opioid receptors also couple to effectors from endosomes activating intracellular enzymes and kinases. In contrast to postsynaptic opioid receptors, receptors localized to presynaptic terminals are resistant to desensitization such that there is no loss of signaling in the continuous presence of opioids over the same time scale. Thus, the balance of opioid signaling in circuits expressing pre- and postsynaptic opioid receptors is shifted toward inhibition of presynaptic neurotransmitter release during continuous opioid exposure. The functional implication of this shift is not often acknowledged in behavioral studies. This review covers what is currently understood about regulation of opioid/nociceptin receptors, with an emphasis on opioid receptor signaling in pain and reward circuits. Importantly, the review covers regulation of presynaptic receptors and the critical gaps in understanding this area, as well as the opportunities to further understand opioid signaling in brain circuits. This article is part of the Special Issue on "Opioid-induced changes in addiction and pain circuits".
Collapse
Affiliation(s)
- Basile Coutens
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Susan L Ingram
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
7
|
Li S, Zhang XQ, Liu CC, Wang ZY, Lu GY, Shen HW, Wu N, Li J, Li F. IRAS/Nischarin modulates morphine reward by glutamate receptor activation in the nucleus accumbens of mouse brain. Biomed Pharmacother 2022; 153:113346. [DOI: 10.1016/j.biopha.2022.113346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/02/2022] Open
|
8
|
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
|
9
|
Buprenorphine: Far Beyond the "Ceiling". Biomolecules 2021; 11:biom11060816. [PMID: 34072706 PMCID: PMC8230089 DOI: 10.3390/biom11060816] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 01/20/2023] Open
Abstract
Chronic pain, including neuropathic pain, represents an untreated disease with important repercussions on the quality of life and huge costs on the national health system. It is well known that opioids are the most powerful analgesic drugs, but they represent the second or third line in neuropathic pain, that remain difficult to manage. Moreover, these drugs show several side effects that limit their use. In addition, opioids possess addictive properties that are associated with misuse and drug abuse. Among available opioids compounds, buprenorphine has been suggested advantageous for a series of clinical reasons, including the effectiveness in neuropathic pain. Some properties are partly explained by its unique pharmacological characteristics. However, questions on the dynamic profile remain to be answered. Pharmacokinetics optimization strategies, and additional potentialities, are still to be explored. In this paper, we attempt to conceptualize the potential undiscovered dynamic profile of buprenorphine.
Collapse
|
10
|
Kudla L, Przewlocki R. Influence of G protein-biased agonists of μ-opioid receptor on addiction-related behaviors. Pharmacol Rep 2021; 73:1033-1051. [PMID: 33835467 PMCID: PMC8413226 DOI: 10.1007/s43440-021-00251-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/05/2021] [Accepted: 03/16/2021] [Indexed: 01/09/2023]
Abstract
Opioid analgesics remain a gold standard for the treatment of moderate to severe pain. However, their clinical utility is seriously limited by a range of adverse effects. Among them, their high-addictive potential appears as very important, especially in the context of the opioid epidemic. Therefore, the development of safer opioid analgesics with low abuse potential appears as a challenging problem for opioid research. Among the last few decades, different approaches to the discovery of novel opioid drugs have been assessed. One of the most promising is the development of G protein-biased opioid agonists, which can activate only selected intracellular signaling pathways. To date, discoveries of several biased agonists acting via μ-opioid receptor were reported. According to the experimental data, such ligands may be devoid of at least some of the opioid side effects, such as respiratory depression or constipation. Nevertheless, most data regarding the addictive properties of biased μ-opioid receptor agonists are inconsistent. A global problem connected with opioid abuse also requires the search for effective pharmacotherapy for opioid addiction, which is another potential application of biased compounds. This review discusses the state-of-the-art on addictive properties of G protein-biased μ-opioid receptor agonists as well as we analyze whether these compounds can diminish any symptoms of opioid addiction. Finally, we provide a critical view on recent data connected with biased signaling and its implications to in vivo manifestations of addiction.
Collapse
Affiliation(s)
- Lucja Kudla
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, ul. Smetna 12, 31-343, Krakow, Poland
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, ul. Smetna 12, 31-343, Krakow, Poland.
| |
Collapse
|
11
|
Sellgren CM, Imbeault S, Larsson MK, Oliveros A, Nilsson IAK, Codeluppi S, Orhan F, Bhat M, Tufvesson-Alm M, Gracias J, Kegel ME, Zheng Y, Faka A, Svedberg M, Powell SB, Caldwell S, Kamenski ME, Vawter MP, Schulmann A, Goiny M, Svensson CI, Hökfelt T, Schalling M, Schwieler L, Cervenka S, Choi DS, Landén M, Engberg G, Erhardt S. GRK3 deficiency elicits brain immune activation and psychosis. Mol Psychiatry 2021; 26:6820-6832. [PMID: 33976392 PMCID: PMC8760053 DOI: 10.1038/s41380-021-01106-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 04/07/2021] [Indexed: 02/03/2023]
Abstract
The G protein-coupled receptor kinase (GRK) family member protein GRK3 has been linked to the pathophysiology of schizophrenia and bipolar disorder. Expression, as well as protein levels, of GRK3 are reduced in post-mortem prefrontal cortex of schizophrenia subjects. Here, we investigate functional behavior and neurotransmission related to immune activation and psychosis using mice lacking functional Grk3 and utilizing a variety of methods, including behavioral, biochemical, electrophysiological, molecular, and imaging methods. Compared to wildtype controls, the Grk3-/- mice show a number of aberrations linked to psychosis, including elevated brain levels of IL-1β, increased turnover of kynurenic acid (KYNA), hyper-responsiveness to D-amphetamine, elevated spontaneous firing of midbrain dopamine neurons, and disruption in prepulse inhibition. Analyzing human genetic data, we observe a link between psychotic features in bipolar disorder, decreased GRK expression, and increased concentration of CSF KYNA. Taken together, our data suggest that Grk3-/- mice show face and construct validity relating to the psychosis phenotype with glial activation and would be suitable for translational studies of novel immunomodulatory agents in psychotic disorders.
Collapse
Affiliation(s)
- Carl M. Sellgren
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden ,grid.4714.60000 0004 1937 0626Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm & Stockholm Health Care Services, Region Stockholm, Sweden
| | - Sophie Imbeault
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Markus K. Larsson
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Alfredo Oliveros
- grid.66875.3a0000 0004 0459 167XDepartment of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN USA
| | - Ida A. K. Nilsson
- grid.4714.60000 0004 1937 0626Translational Psychiatry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Simone Codeluppi
- grid.4714.60000 0004 1937 0626Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Funda Orhan
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Bhat
- grid.418151.80000 0001 1519 6403Research and Development, Innovative Medicines, Personalised Healthcare and Biomarkers, Translational Science Centre, Science for Life Laboratory, AstraZeneca, Solna, Sweden ,grid.4714.60000 0004 1937 0626Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Maximilian Tufvesson-Alm
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jessica Gracias
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Magdalena E. Kegel
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Yiran Zheng
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anthi Faka
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Svedberg
- grid.4714.60000 0004 1937 0626Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Susan B. Powell
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Sorana Caldwell
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Mary E. Kamenski
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Marquis P. Vawter
- grid.266093.80000 0001 0668 7243Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine School of Medicine, Irvine, CA USA
| | - Anton Schulmann
- grid.416868.50000 0004 0464 0574Human Genetics Branch, National Institute of Mental Health, Bethesda, MD USA
| | - Michel Goiny
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Camilla I. Svensson
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Hökfelt
- grid.4714.60000 0004 1937 0626Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Schalling
- grid.4714.60000 0004 1937 0626Translational Psychiatry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lilly Schwieler
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Simon Cervenka
- grid.4714.60000 0004 1937 0626Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm & Stockholm Health Care Services, Region Stockholm, Sweden
| | - Doo-Sup Choi
- grid.66875.3a0000 0004 0459 167XDepartment of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN USA ,grid.66875.3a0000 0004 0459 167XDepartment of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN USA
| | - Mikael Landén
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden ,grid.4714.60000 0004 1937 0626Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Göran Engberg
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | - Sophie Erhardt
- grid.4714.60000 0004 1937 0626Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Gurevich EV, Gurevich VV. GRKs as Modulators of Neurotransmitter Receptors. Cells 2020; 10:cells10010052. [PMID: 33396400 PMCID: PMC7823573 DOI: 10.3390/cells10010052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 01/08/2023] Open
Abstract
Many receptors for neurotransmitters, such as dopamine, norepinephrine, acetylcholine, and neuropeptides, belong to the superfamily of G protein-coupled receptors (GPCRs). A general model posits that GPCRs undergo two-step homologous desensitization: the active receptor is phosphorylated by kinases of the G protein-coupled receptor kinase (GRK) family, whereupon arrestin proteins specifically bind active phosphorylated receptors, shutting down G protein-mediated signaling, facilitating receptor internalization, and initiating distinct signaling pathways via arrestin-based scaffolding. Here, we review the mechanisms of GRK-dependent regulation of neurotransmitter receptors, focusing on the diverse modes of GRK-mediated phosphorylation of receptor subtypes. The immediate signaling consequences of GRK-mediated receptor phosphorylation, such as arrestin recruitment, desensitization, and internalization/resensitization, are equally diverse, depending not only on the receptor subtype but also on phosphorylation by GRKs of select receptor residues. We discuss the signaling outcome as well as the biological and behavioral consequences of the GRK-dependent phosphorylation of neurotransmitter receptors where known.
Collapse
|
13
|
GRKs as Key Modulators of Opioid Receptor Function. Cells 2020; 9:cells9112400. [PMID: 33147802 PMCID: PMC7692057 DOI: 10.3390/cells9112400] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/21/2020] [Accepted: 10/28/2020] [Indexed: 12/21/2022] Open
Abstract
Understanding the link between agonist-induced phosphorylation of the mu-opioid receptor (MOR) and the associated physiological effects is critical for the development of novel analgesic drugs and is particularly important for understanding the mechanisms responsible for opioid-induced tolerance and addiction. The family of G protein receptor kinases (GRKs) play a pivotal role in such processes, mediating phosphorylation of residues at the C-tail of opioid receptors. Numerous strategies, such as phosphosite specific antibodies and mass spectrometry have allowed the detection of phosphorylated residues and the use of mutant knock-in mice have shed light on the role of GRK regulation in opioid receptor physiology. Here we review our current understanding on the role of GRKs in the actions of opioid receptors, with a particular focus on the MOR, the target of most commonly used opioid analgesics such as morphine or fentanyl.
Collapse
|
14
|
Møller TC, Pedersen MF, van Senten JR, Seiersen SD, Mathiesen JM, Bouvier M, Bräuner-Osborne H. Dissecting the roles of GRK2 and GRK3 in μ-opioid receptor internalization and β-arrestin2 recruitment using CRISPR/Cas9-edited HEK293 cells. Sci Rep 2020; 10:17395. [PMID: 33060647 PMCID: PMC7567791 DOI: 10.1038/s41598-020-73674-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 09/18/2020] [Indexed: 01/14/2023] Open
Abstract
Most G protein-coupled receptors (GPCRs) recruit β-arrestins and internalize upon agonist stimulation. For the μ-opioid receptor (μ-OR), this process has been linked to development of opioid tolerance. GPCR kinases (GRKs), particularly GRK2 and GRK3, have been shown to be important for μ-OR recruitment of β-arrestin and internalization. However, the contribution of GRK2 and GRK3 to β-arrestin recruitment and receptor internalization, remain to be determined in their complete absence. Using CRISPR/Cas9-mediated genome editing we established HEK293 cells with knockout of GRK2, GRK3 or both to dissect their individual contributions in β-arrestin2 recruitment and μ-OR internalization upon stimulation with four different agonists. We showed that GRK2/3 removal reduced agonist-induced μ-OR internalization and β-arrestin2 recruitment substantially and we found GRK2 to be more important for these processes than GRK3. Furthermore, we observed a sustained and GRK2/3 independent component of β-arrestin2 recruitment to the plasma membrane upon μ-OR activation. Rescue expression experiments restored GRK2/3 functions. Inhibition of GRK2/3 using the small molecule inhibitor CMPD101 showed a high similarity between the genetic and pharmacological approaches, cross-validating the specificity of both. However, off-target effects were observed at high CMPD101 concentrations. These GRK2/3 KO cell lines should prove useful for a wide range of studies on GPCR function.
Collapse
Affiliation(s)
- Thor C Møller
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark.
| | - Mie F Pedersen
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Jeffrey R van Senten
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Sofie D Seiersen
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Jesper M Mathiesen
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark.
| |
Collapse
|
15
|
Lemos Duarte M, Devi LA. Post-translational Modifications of Opioid Receptors. Trends Neurosci 2020; 43:417-432. [PMID: 32459993 PMCID: PMC7323054 DOI: 10.1016/j.tins.2020.03.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 12/13/2022]
Abstract
Post-translational modifications (PTMs) are key events in signal transduction since they affect protein function by regulating their abundance and/or activity. PTMs involve the covalent attachment of functional groups to specific amino acids. Since they tend to be generally reversible, PTMs serve as regulators of signal transduction pathways. G-protein-coupled receptors (GPCRs) are major signaling proteins that undergo multiple types of PTMs. In this Review, we focus on the opioid receptors, members of GPCR family A, and highlight recent advances in the field that have underscored the importance of PTMs in the functional regulation of these receptors. Since opioid receptor activity plays a central role in the development of tolerance and addiction to morphine and other drugs of abuse, understanding the molecular mechanisms regulating receptor activity is of fundamental importance.
Collapse
Affiliation(s)
- Mariana Lemos Duarte
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
16
|
Agonist-induced phosphorylation bar code and differential post-activation signaling of the delta opioid receptor revealed by phosphosite-specific antibodies. Sci Rep 2020; 10:8585. [PMID: 32444688 PMCID: PMC7244497 DOI: 10.1038/s41598-020-65589-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/05/2020] [Indexed: 01/08/2023] Open
Abstract
The δ-opioid receptor (DOP) is an attractive pharmacological target due to its potent analgesic, anxiolytic and anti-depressant activity in chronic pain models. However, some but not all selective DOP agonists also produce severe adverse effects such as seizures. Thus, the development of novel agonists requires a profound understanding of their effects on DOP phosphorylation, post-activation signaling and dephosphorylation. Here we show that agonist-induced DOP phosphorylation at threonine 361 (T361) and serine 363 (S363) proceeds with a temporal hierarchy, with S363 as primary site of phosphorylation. This phosphorylation is mediated by G protein-coupled receptor kinases 2 and 3 (GRK2/3) followed by DOP endocytosis and desensitization. DOP dephosphorylation occurs within minutes and is predominantly mediated by protein phosphatases (PP) 1α and 1β. A comparison of structurally diverse DOP agonists and clinically used opioids demonstrated high correlation between G protein-dependent signaling efficacies and receptor internalization. In vivo, DOP agonists induce receptor phosphorylation in a dose-dependent and agonist-selective manner that could be blocked by naltrexone in DOP-eGFP mice. Together, our studies provide novel tools and insights for ligand-activated DOP signaling in vitro and in vivo and suggest that DOP agonist efficacies may determine receptor post-activation signaling.
Collapse
|
17
|
Fernandez TJ, De Maria M, Lobingier BT. A cellular perspective of bias at G protein-coupled receptors. Protein Sci 2020; 29:1345-1354. [PMID: 32297394 DOI: 10.1002/pro.3872] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/17/2022]
Abstract
G protein-coupled receptors (GPCRs) modulate cell function over short- and long-term timescales. GPCR signaling depends on biochemical parameters that define the what, when, and where of receptor function: what proteins mediate and regulate receptor signaling, where within the cell these interactions occur, and how long these interactions persist. These parameters can vary significantly depending on the activating ligand. Collectivity, differential agonist activity at a GPCR is called bias or functional selectivity. Here we review agonist bias at GPCRs with a focus on ligands that show dramatically different cellular responses from their unbiased counterparts.
Collapse
Affiliation(s)
- Thomas J Fernandez
- Department of Chemical Physiology and Biochemistry, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Monica De Maria
- Department of Chemical Physiology and Biochemistry, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Braden T Lobingier
- Department of Chemical Physiology and Biochemistry, Oregon Health and Sciences University, Portland, Oregon, USA
| |
Collapse
|
18
|
Bouley RA, Weinberg ZY, Waldschmidt HV, Yen YC, Larsen SD, Puthenveedu MA, Tesmer JJG. A New Paroxetine-Based GRK2 Inhibitor Reduces Internalization of the μ-Opioid Receptor. Mol Pharmacol 2020; 97:392-401. [PMID: 32234810 DOI: 10.1124/mol.119.118661] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
G protein-coupled receptor (GPCR) kinases (GRKs) play a key role in terminating signals initiated by agonist-bound GPCRs. However, chronic stimulation of GPCRs, such as that which occurs during heart failure, leads to the overexpression of GRKs and maladaptive downregulation of GPCRs on the cell surface. We previously reported the discovery of potent and selective families of GRK inhibitors based on either the paroxetine or GSK180736A scaffold. A new inhibitor, CCG258747, which is based on paroxetine, demonstrates increased potency against the GRK2 subfamily and favorable pharmacokinetic parameters in mice. CCG258747 and the closely related compound CCG258208 also showed high selectivity for the GRK2 subfamily in a kinome panel of 104 kinases. We developed a cell-based assay to screen the ability of CCG258747 and 10 other inhibitors with different GRK subfamily selectivities and with either the paroxetine or GSK180736A scaffold to block internalization of the μ-opioid receptor (MOR). CCG258747 showed the best efficacy in blocking MOR internalization among the compounds tested. Furthermore, we show that compounds based on paroxetine had much better cell permeability than those based on GSK180736A, which explains why GSK180736A-based inhibitors, although being potent in vitro, do not always show efficacy in cell-based assays. This study validates the paroxetine scaffold as the most effective for GRK inhibition in living cells, confirming that GRK2 predominantly drives internalization of MOR in the cell lines we tested and underscores the utility of high-resolution cell-based assays for assessment of compound efficacy. SIGNIFICANCE STATEMENT: G protein-coupled receptor kinases (GRKs) are attractive targets for developing therapeutics for heart failure. We have synthesized a new GRK2 subfamily-selective inhibitor, CCG258747, which has nanomolar potency against GRK2 and excellent selectivity over other kinases. A live-cell receptor internalization assay was used to test the ability of GRK2 inhibitors to impart efficacy on a GRK-dependent process in cells. Our data indicate that CCG258747 blocked the internalization of the μ-opioid receptor most efficaciously because it has the ability to cross cell membranes.
Collapse
Affiliation(s)
- Renee A Bouley
- Life Sciences Institute (R.A.B., H.V.W.), Departments of Medicinal Chemistry (H.V.W., S.D.L.) and Pharmacology (R.A.B., Z.Y.W., M.A.P.), and Vahlteich Medicinal Chemistry Core, College of Pharmacy (H.V.W., S.D.L.), University of Michigan, Ann Arbor, Michigan; and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology (Y.-C.Y., J.J.G.T.), Purdue University, West Lafayette, Indiana
| | - Zara Y Weinberg
- Life Sciences Institute (R.A.B., H.V.W.), Departments of Medicinal Chemistry (H.V.W., S.D.L.) and Pharmacology (R.A.B., Z.Y.W., M.A.P.), and Vahlteich Medicinal Chemistry Core, College of Pharmacy (H.V.W., S.D.L.), University of Michigan, Ann Arbor, Michigan; and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology (Y.-C.Y., J.J.G.T.), Purdue University, West Lafayette, Indiana
| | - Helen V Waldschmidt
- Life Sciences Institute (R.A.B., H.V.W.), Departments of Medicinal Chemistry (H.V.W., S.D.L.) and Pharmacology (R.A.B., Z.Y.W., M.A.P.), and Vahlteich Medicinal Chemistry Core, College of Pharmacy (H.V.W., S.D.L.), University of Michigan, Ann Arbor, Michigan; and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology (Y.-C.Y., J.J.G.T.), Purdue University, West Lafayette, Indiana
| | - Yu-Chen Yen
- Life Sciences Institute (R.A.B., H.V.W.), Departments of Medicinal Chemistry (H.V.W., S.D.L.) and Pharmacology (R.A.B., Z.Y.W., M.A.P.), and Vahlteich Medicinal Chemistry Core, College of Pharmacy (H.V.W., S.D.L.), University of Michigan, Ann Arbor, Michigan; and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology (Y.-C.Y., J.J.G.T.), Purdue University, West Lafayette, Indiana
| | - Scott D Larsen
- Life Sciences Institute (R.A.B., H.V.W.), Departments of Medicinal Chemistry (H.V.W., S.D.L.) and Pharmacology (R.A.B., Z.Y.W., M.A.P.), and Vahlteich Medicinal Chemistry Core, College of Pharmacy (H.V.W., S.D.L.), University of Michigan, Ann Arbor, Michigan; and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology (Y.-C.Y., J.J.G.T.), Purdue University, West Lafayette, Indiana
| | - Manojkumar A Puthenveedu
- Life Sciences Institute (R.A.B., H.V.W.), Departments of Medicinal Chemistry (H.V.W., S.D.L.) and Pharmacology (R.A.B., Z.Y.W., M.A.P.), and Vahlteich Medicinal Chemistry Core, College of Pharmacy (H.V.W., S.D.L.), University of Michigan, Ann Arbor, Michigan; and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology (Y.-C.Y., J.J.G.T.), Purdue University, West Lafayette, Indiana
| | - John J G Tesmer
- Life Sciences Institute (R.A.B., H.V.W.), Departments of Medicinal Chemistry (H.V.W., S.D.L.) and Pharmacology (R.A.B., Z.Y.W., M.A.P.), and Vahlteich Medicinal Chemistry Core, College of Pharmacy (H.V.W., S.D.L.), University of Michigan, Ann Arbor, Michigan; and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology (Y.-C.Y., J.J.G.T.), Purdue University, West Lafayette, Indiana
| |
Collapse
|
19
|
Grim TW, Acevedo-Canabal A, Bohn LM. Toward Directing Opioid Receptor Signaling to Refine Opioid Therapeutics. Biol Psychiatry 2020; 87:15-21. [PMID: 31806082 PMCID: PMC6919561 DOI: 10.1016/j.biopsych.2019.10.020] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/22/2022]
Abstract
The mu opioid receptor (MOR) is a diversely regulated target for the alleviation of pain in the clinical setting. However, untoward side effects such as tolerance, dependence, respiratory suppression, constipation, and abuse liability detract from the general activation of these receptors. Studies in genetically modified rodent models suggest that activating G protein signaling pathways while avoiding phosphorylation of the receptor or recruitment of β-arrestin scaffolding proteins could preserve the analgesic properties of MOR agonists while avoiding certain side effects. With the development of novel MOR "biased" agonists, which lead to preferential activation of G protein pathways over receptor phosphorylation, internalization, or interaction with other effectors, this hypothesis can be tested in a native, physiological setting. Overall, it is clear that the MOR is not a simple on-off switch and that the diverse means by which the receptor can be regulated may present an opportunity to refine therapeutics for the treatment of pain.
Collapse
Affiliation(s)
- Travis W Grim
- Departments of Molecular Medicine and Neuroscience, the Scripps Research Institute, Jupiter, Florida
| | - Agnes Acevedo-Canabal
- Departments of Molecular Medicine and Neuroscience, the Scripps Research Institute, Jupiter, Florida
| | - Laura M Bohn
- Departments of Molecular Medicine and Neuroscience, the Scripps Research Institute, Jupiter, Florida.
| |
Collapse
|
20
|
Pedersen MF, Wróbel TM, Märcher-Rørsted E, Pedersen DS, Møller TC, Gabriele F, Pedersen H, Matosiuk D, Foster SR, Bouvier M, Bräuner-Osborne H. Biased agonism of clinically approved μ-opioid receptor agonists and TRV130 is not controlled by binding and signaling kinetics. Neuropharmacology 2019; 166:107718. [PMID: 31351108 DOI: 10.1016/j.neuropharm.2019.107718] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/08/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022]
Abstract
Binding and signaling kinetics have previously proven important in validation of biased agonism at GPCRs. Here we provide a comprehensive kinetic pharmacological comparison of clinically relevant μ-opioid receptor agonists, including the novel biased agonist oliceridine (TRV130) which is in clinical trial for pain management. We demonstrate that the bias profile observed for the selected agonists is not time-dependent and that agonists with dramatic differences in their binding kinetic properties can display the same degree of bias. Binding kinetics analyses demonstrate that buprenorphine has 18-fold higher receptor residence time than oliceridine. This is thus the largest pharmacodynamic difference between the clinically approved drug buprenorphine and the clinical candidate oliceridine, since their bias profiles are similar. Further, we provide the first pharmacological characterization of (S)-TRV130 demonstrating that it has a similar pharmacological profile as the (R)-form, oliceridine, but displays 90-fold lower potency than the (R)-form. This difference is driven by a significantly slower association rate. Finally, we show that the selected agonists are differentially affected by G protein-coupled receptor kinase 2 and 5 (GRK2 and GRK5) expression. GRK2 and GRK5 overexpression greatly increased μ-opioid receptor internalization induced by morphine, but only had modest effects on buprenorphine and oliceridine-induced internalization. Overall, our data reveal that the clinically available drug buprenorphine displays a similar pharmacological bias profile in vitro compared to the clinical candidate drug oliceridine and that this bias is independent of binding kinetics suggesting a mechanism driven by receptor-conformations. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.
Collapse
Affiliation(s)
- Mie Fabricius Pedersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark; Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
| | - Tomasz Marcin Wróbel
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark; Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Medical University of Lublin, Lublin, Poland
| | - Emil Märcher-Rørsted
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Sejer Pedersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Thor Christian Møller
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Federica Gabriele
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | - Dariusz Matosiuk
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Medical University of Lublin, Lublin, Poland
| | - Simon Richard Foster
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, Canada.
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
21
|
Mann A, Moulédous L, Froment C, O'Neill PR, Dasgupta P, Günther T, Brunori G, Kieffer BL, Toll L, Bruchas MR, Zaveri NT, Schulz S. Agonist-selective NOP receptor phosphorylation correlates in vitro and in vivo and reveals differential post-activation signaling by chemically diverse agonists. Sci Signal 2019; 12:12/574/eaau8072. [PMID: 30914485 DOI: 10.1126/scisignal.aau8072] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Agonists of the nociceptin/orphanin FQ opioid peptide (NOP) receptor, a member of the opioid receptor family, are under active investigation as novel analgesics, but their modes of signaling are less well characterized than those of other members of the opioid receptor family. Therefore, we investigated whether different NOP receptor ligands showed differential signaling or functional selectivity at the NOP receptor. Using newly developed phosphosite-specific antibodies to the NOP receptor, we found that agonist-induced NOP receptor phosphorylation occurred primarily at four carboxyl-terminal serine (Ser) and threonine (Thr) residues, namely, Ser346, Ser351, Thr362, and Ser363, and proceeded with a temporal hierarchy, with Ser346 as the first site of phosphorylation. G protein-coupled receptor kinases 2 and 3 (GRK2/3) cooperated during agonist-induced phosphorylation, which, in turn, facilitated NOP receptor desensitization and internalization. A comparison of structurally distinct NOP receptor agonists revealed dissociation in functional efficacies between G protein-dependent signaling and receptor phosphorylation. Furthermore, in NOP-eGFP and NOP-eYFP mice, NOP receptor agonists induced multisite phosphorylation and internalization in a dose-dependent and agonist-selective manner that could be blocked by specific antagonists. Our study provides new tools to study ligand-activated NOP receptor signaling in vitro and in vivo. Differential agonist-selective NOP receptor phosphorylation by chemically diverse NOP receptor agonists suggests that differential signaling by NOP receptor agonists may play a role in NOP receptor ligand pharmacology.
Collapse
Affiliation(s)
- Anika Mann
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany.
| | - Lionel Moulédous
- Research Center on Animal Cognition, Center for Integrative Biology, Toulouse University, CNRS, UPS, 31062 Toulouse Cedex 09, France
| | - Carine Froment
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, 31077 Toulouse Cedex 04, France
| | - Patrick R O'Neill
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Pooja Dasgupta
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany
| | - Thomas Günther
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany
| | - Gloria Brunori
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Brigitte L Kieffer
- Douglas Research Center, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC H3A 1A1, Canada
| | - Lawrence Toll
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Michael R Bruchas
- Center for the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pharmacology, University of Washington, Seattle, WA 98195, USA
| | | | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany.
| |
Collapse
|
22
|
Kliewer A, Schmiedel F, Sianati S, Bailey A, Bateman JT, Levitt ES, Williams JT, Christie MJ, Schulz S. Phosphorylation-deficient G-protein-biased μ-opioid receptors improve analgesia and diminish tolerance but worsen opioid side effects. Nat Commun 2019; 10:367. [PMID: 30664663 PMCID: PMC6341117 DOI: 10.1038/s41467-018-08162-1] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022] Open
Abstract
Opioid analgesics are powerful pain relievers; however, over time, pain control diminishes as analgesic tolerance develops. The molecular mechanisms initiating tolerance have remained unresolved to date. We have previously shown that desensitization of the μ-opioid receptor and interaction with β-arrestins is controlled by carboxyl-terminal phosphorylation. Here we created knockin mice with a series of serine- and threonine-to-alanine mutations that render the receptor increasingly unable to recruit β-arrestins. Desensitization is inhibited in locus coeruleus neurons of mutant mice. Opioid-induced analgesia is strongly enhanced and analgesic tolerance is greatly diminished. Surprisingly, respiratory depression, constipation, and opioid withdrawal signs are unchanged or exacerbated, indicating that β-arrestin recruitment does not contribute to the severity of opioid side effects and, hence, predicting that G-protein-biased µ-agonists are still likely to elicit severe adverse effects. In conclusion, our findings identify carboxyl-terminal multisite phosphorylation as key step that drives acute μ-opioid receptor desensitization and long-term tolerance.
Collapse
Affiliation(s)
- A Kliewer
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, 07747, Jena, Germany
| | - F Schmiedel
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, 07747, Jena, Germany
| | - S Sianati
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, NSW, 2006, Australia
| | - A Bailey
- Institute of Medical and Biomedical Education, St George's University of London, London, SW17 ORE, UK
| | - J T Bateman
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, 32608, USA
| | - E S Levitt
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, 32608, USA
| | - J T Williams
- The Vollum Institute, Oregon Health and Science University, 3181S.W. Sam Jackson Pk. Rd., Portland, OR, 97239, USA
| | - M J Christie
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, NSW, 2006, Australia
| | - S Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, 07747, Jena, Germany.
| |
Collapse
|
23
|
Pasternak GW, Childers SR, Pan YX. Emerging Insights into Mu Opioid Pharmacology. Handb Exp Pharmacol 2019; 258:89-125. [PMID: 31598835 DOI: 10.1007/164_2019_270] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Opioid analgesics, most of which act through mu opioid receptors, have long represented valuable therapeutic agents to treat severe pain. Concerted drug development efforts for over a 100 years have resulted in a large variety of opioid analgesics used in the clinic, but all of them continue to exhibit the side effects, especially respiratory depression, that have long plagued the use of morphine. The recent explosion in fatalities resulting from overdose of prescription and synthetic opioids has dramatically increased the need for safer analgesics, but recent developments in mu receptor research have provided new strategies to develop such drugs. This chapter reviews recent advances in developing novel opioid analgesics from an understanding of mu receptor structure and function. This includes a summary of the mechanism of agonist binding deduced from the crystal structure of mu receptors. It will also highlight the development of novel agonist mechanisms, including biased agonists, bivalent ligands, and allosteric modulators of mu receptor function, and describe how receptor phosphorylation modulates these pathways. Finally, it will summarize research on the alternative pre-mRNA splicing mechanisms that produces a multiplicity of mu receptor isoforms. Many of these isoforms exhibit different pharmacological specificities and brain circuitry localization, thus providing an opportunity to develop novel drugs with increased therapeutic windows.
Collapse
Affiliation(s)
- Gavril W Pasternak
- Department of Neurology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Steven R Childers
- Department of Physiology/Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - Ying-Xian Pan
- Department of Neurology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
24
|
Wang SC, Chung RH, Kuo HW, Liu TH, Fang CP, Liu SC, Liu CC, Tsou HH, Chen ACH, Liu YL. GRK5 Is Associated with the Regulation of Methadone Dosage in Heroin Dependence. Int J Neuropsychopharmacol 2018; 21:910-917. [PMID: 30060048 PMCID: PMC6165957 DOI: 10.1093/ijnp/pyy066] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/24/2018] [Accepted: 07/25/2018] [Indexed: 12/25/2022] Open
Abstract
Background There is no countable biomarker for opioid dependence treatment responses thus far. In this study, we recruited Taiwanese methadone maintenance treatment patients to search for genes involving the regulatory mechanisms of methadone dose by genome-wide association analyses. Methods A total of 344 Taiwanese methadone maintenance treatment patients were included in a genome-wide association study. The involvement of GRK5 in opioid dependence was then further confirmed by gene expression study on lymphoblastoid cell lines derived from 3 independent age- and gender-matched groups: methadone maintenance treatment patients, medication-free former heroin abusers, and normal controls. Results The results indicated that GRK5, the gene encoding an enzyme related to μ-opioid receptor desensitization, is associated with methadone dose by additive model of gene-based association analysis (P=6.76×10-5). We found that 6 of the 55 single nucleotide polymorphisms from the genome-wide genotype platform and 2 single nucleotide polymorphisms from the 29 additionally selected single nucleotide polymorphisms were significantly associated with methadone maintenance dose in both genotype and allele type (P ≤ .006), especially in patients who tested negative in the urine morphine test. The levels of GRK5 gene expression were similar between methadone maintenance treatment patients and medication-free former heroin abusers. However, the normal controls showed a significantly lower level of GRK5 gene expression than the other groups (P=.019). Conclusions The results suggested an important role for GRK5 in the regulatory mechanisms of methadone dose and course of heroin dependence.
Collapse
Affiliation(s)
- Sheng-Chang Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Ren-Hua Chung
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Hsiang-Wei Kuo
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Tung-Hsia Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Chiu-Ping Fang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Shu Chih Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Chia-Chen Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Hsiao-Hui Tsou
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
- Graduate Institute of Biostatistics, College of Public Health, China Medical University, Taichung, Taiwan
| | - Andrew C H Chen
- Department of Psychiatry, the Zucker Hillside Hospital, Northwell Health, Glen Oaks, New York
- The Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine at Hofstra University, Manhasset, New York
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
25
|
Miess E, Gondin AB, Yousuf A, Steinborn R, Mösslein N, Yang Y, Göldner M, Ruland JG, Bünemann M, Krasel C, Christie MJ, Halls ML, Schulz S, Canals M. Multisite phosphorylation is required for sustained interaction with GRKs and arrestins during rapid μ-opioid receptor desensitization. Sci Signal 2018; 11:11/539/eaas9609. [PMID: 30018083 DOI: 10.1126/scisignal.aas9609] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
G protein receptor kinases (GRKs) and β-arrestins are key regulators of μ-opioid receptor (MOR) signaling and trafficking. We have previously shown that high-efficacy opioids such as DAMGO stimulate a GRK2/3-mediated multisite phosphorylation of conserved C-terminal tail serine and threonine residues, which facilitates internalization of the receptor. In contrast, morphine-induced phosphorylation of MOR is limited to Ser375 and is not sufficient to drive substantial receptor internalization. We report how specific multisite phosphorylation controlled the dynamics of GRK and β-arrestin interactions with MOR and show how such phosphorylation mediated receptor desensitization. We showed that GRK2/3 was recruited more quickly than was β-arrestin to a DAMGO-activated MOR. β-Arrestin recruitment required GRK2 activity and MOR phosphorylation, but GRK recruitment also depended on the phosphorylation sites in the C-terminal tail, specifically four serine and threonine residues within the 370TREHPSTANT379 motif. Our results also suggested that other residues outside this motif participated in the initial and transient recruitment of GRK and β-arrestins. We identified two components of high-efficacy agonist desensitization of MOR: a sustained component, which required GRK2-mediated phosphorylation and a potential soluble factor, and a rapid component, which was likely mediated by GRK2 but independent of receptor phosphorylation. Elucidating these complex receptor-effector interactions represents an important step toward a mechanistic understanding of MOR desensitization that leads to the development of tolerance and dependence.
Collapse
Affiliation(s)
- Elke Miess
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, D-07747 Jena, Germany
| | - Arisbel B Gondin
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Victoria 3052, Australia
| | - Arsalan Yousuf
- Discipline of Pharmacology, University of Sydney, New South Wales 2006, Australia
| | - Ralph Steinborn
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, D-07747 Jena, Germany
| | - Nadja Mösslein
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Yunshi Yang
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Martin Göldner
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Julia G Ruland
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Moritz Bünemann
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Cornelius Krasel
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - MacDonald J Christie
- Discipline of Pharmacology, University of Sydney, New South Wales 2006, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Victoria 3052, Australia
| | - Stefan Schulz
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, D-07747 Jena, Germany.
| | - Meritxell Canals
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Victoria 3052, Australia.
| |
Collapse
|
26
|
Caputi FF, Acquas E, Kasture S, Ruiu S, Candeletti S, Romualdi P. The standardized Withania somnifera Dunal root extract alters basal and morphine-induced opioid receptor gene expression changes in neuroblastoma cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:9. [PMID: 29316911 PMCID: PMC5761194 DOI: 10.1186/s12906-017-2065-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/15/2017] [Indexed: 12/17/2022]
Abstract
Background Behavioral studies demonstrated that the administration of Withania somnifera Dunal roots extract (WSE), prolongs morphine-elicited analgesia and reduces the development of tolerance to the morphine’s analgesic effect; however, little is known about the underpinning molecular mechanism(s). In order to shed light on this issue in the present paper we explored whether WSE promotes alterations of μ (MOP) and nociceptin (NOP) opioid receptors gene expression in neuroblastoma SH-SY5Y cells. Methods A range of WSE concentrations was preliminarily tested to evaluate their effects on cell viability. Subsequently, the effects of 5 h exposure to WSE (0.25, 0.50 and 1.00 mg/ml), applied alone and in combination with morphine or naloxone, on MOP and NOP mRNA levels were investigated. Results Data analysis revealed that morphine decreased MOP and NOP receptor gene expression, whereas naloxone elicited their up-regulation. In addition, pre-treatment with naloxone prevented the morphine-elicited gene expression alterations. Interestingly, WSE was able to: a) alter MOP but not NOP gene expression; b) counteract, at its highest concentration, morphine-induced MOP down-regulation, and c) hamper naloxone-induced MOP and NOP up-regulation. Conclusion Present in-vitro data disclose novel evidence about the ability of WSE to influence MOP and NOP opioid receptors gene expression in SH-SY5Y cells. Moreover, our findings suggest that the in-vivo modulation of morphine-mediated analgesia by WSE could be related to the hindering of morphine-elicited opioid receptors down-regulation here observed following WSE pre-treatment at its highest concentration.
Collapse
|
27
|
Abstract
Opioids are powerful analgesics, but also carry significant side effects and abuse potential. Here we describe a modulator of the μ-opioid receptor (MOR1), the transient receptor potential channel subfamily vanilloid member 1 (TRPV1). We show that TRPV1 binds MOR1 and blocks opioid-dependent phosphorylation of MOR1 while leaving G protein signaling intact. Phosphorylation of MOR1 initiates recruitment and activation of the β-arrestin pathway, which is responsible for numerous opioid-induced adverse effects, including the development of tolerance and respiratory depression. Phosphorylation stands in contrast to G protein signaling, which is responsible for the analgesic effect of opioids. Calcium influx through TRPV1 causes a calcium/calmodulin-dependent translocation of G protein-coupled receptor kinase 5 (GRK5) away from the plasma membrane, thereby blocking its ability to phosphorylate MOR1. Using TRPV1 to block phosphorylation of MOR1 without affecting G protein signaling is a potential strategy to improve the therapeutic profile of opioids.
Collapse
|
28
|
Emerging Paradigms of G Protein-Coupled Receptor Dephosphorylation. Trends Pharmacol Sci 2017; 38:621-636. [DOI: 10.1016/j.tips.2017.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022]
|
29
|
Abstract
This paper is the thirty-eighth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2015 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
30
|
Nakamura A, Ono H, Ando A, Hinata M, Niidome K, Omachi S, Sakaguchi G, Shinohara S. Suppression of the acute upregulation of phosphorylated-extracellular regulated kinase in ventral tegmental area by a μ-opioid receptor agonist is related to resistance to rewarding effects in a mouse model of bone cancer. J Pharmacol Sci 2016; 133:9-17. [PMID: 28034513 DOI: 10.1016/j.jphs.2016.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 09/09/2016] [Accepted: 11/13/2016] [Indexed: 10/20/2022] Open
Abstract
We investigated the mechanisms underlying the suppression of the rewarding effects of opioids using the femur bone cancer (FBC) mouse model. The rewarding and antinociceptive effects of subcutaneously administered morphine and oxycodone in the FBC model mice were assessed using the conditioned place preference test and the von-Frey test. In FBC mice, antinociceptive doses of morphine (30 mg/kg) and oxycodone (5 mg/kg) did not produce the rewarding effects but excessive doses of morphine (300 mg/kg) and oxycodone (100 mg/kg) did. Western blot analyses revealed a transient and significant increase in phosphorylated-extracellular regulated kinase (p-ERK) levels in ventral tegmental area (VTA) 5 min after the administration of morphine in sham-group. Interestingly, in FBC group, a regular dose of morphine did not increase p-ERK levels but a high dose of morphine caused an increase in p-ERK level 5 min after administration. The rewarding effects of a regular dose of and a high dose of morphine in the sham-operation and FBC model, respectively, were significantly inhibited by the MEK inhibitor. The suppression of p-ERK might result in resistance to these rewarding effects under the conditions of bone cancer.
Collapse
Affiliation(s)
- Atsushi Nakamura
- Pain & Neuroscience, Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., 1-1, 3-chome, Futaba-cho, Toyonaka, Osaka 561-0825, Japan.
| | - Hiroko Ono
- Pain & Neuroscience, Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., 1-1, 3-chome, Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Azusa Ando
- Pain & Neuroscience, Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., 1-1, 3-chome, Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Mikie Hinata
- Pain & Neuroscience, Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., 1-1, 3-chome, Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Kazuki Niidome
- Pain & Neuroscience, Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., 1-1, 3-chome, Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Shigeki Omachi
- Pain & Neuroscience, Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., 1-1, 3-chome, Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Gaku Sakaguchi
- Pain & Neuroscience, Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., 1-1, 3-chome, Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Shunji Shinohara
- Pain & Neuroscience, Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., 1-1, 3-chome, Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| |
Collapse
|
31
|
Toll L, Bruchas MR, Calo' G, Cox BM, Zaveri NT. Nociceptin/Orphanin FQ Receptor Structure, Signaling, Ligands, Functions, and Interactions with Opioid Systems. Pharmacol Rev 2016; 68:419-57. [PMID: 26956246 PMCID: PMC4813427 DOI: 10.1124/pr.114.009209] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The NOP receptor (nociceptin/orphanin FQ opioid peptide receptor) is the most recently discovered member of the opioid receptor family and, together with its endogenous ligand, N/OFQ, make up the fourth members of the opioid receptor and opioid peptide family. Because of its more recent discovery, an understanding of the cellular and behavioral actions induced by NOP receptor activation are less well developed than for the other members of the opioid receptor family. All of these factors are important because NOP receptor activation has a clear modulatory role on mu opioid receptor-mediated actions and thereby affects opioid analgesia, tolerance development, and reward. In addition to opioid modulatory actions, NOP receptor activation has important effects on motor function and other physiologic processes. This review discusses how NOP pharmacology intersects, contrasts, and interacts with the mu opioid receptor in terms of tertiary structure and mechanism of receptor activation; location of receptors in the central nervous system; mechanisms of desensitization and downregulation; cellular actions; intracellular signal transduction pathways; and behavioral actions with respect to analgesia, tolerance, dependence, and reward. This is followed by a discussion of the agonists and antagonists that have most contributed to our current knowledge. Because NOP receptors are highly expressed in brain and spinal cord and NOP receptor activation sometimes synergizes with mu receptor-mediated actions and sometimes opposes them, an understanding of NOP receptor pharmacology in the context of these interactions with the opioid receptors will be crucial to the development of novel therapeutics that engage the NOP receptor.
Collapse
Affiliation(s)
- Lawrence Toll
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Michael R Bruchas
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Girolamo Calo'
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Brian M Cox
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Nurulain T Zaveri
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| |
Collapse
|
32
|
Mambretti EM, Kistner K, Mayer S, Massotte D, Kieffer BL, Hoffmann C, Reeh PW, Brack A, Asan E, Rittner HL. Functional and structural characterization of axonal opioid receptors as targets for analgesia. Mol Pain 2016; 12:12/0/1744806916628734. [PMID: 27030709 PMCID: PMC4994859 DOI: 10.1177/1744806916628734] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/10/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Opioids are the gold standard for the treatment of acute pain despite serious side effects in the central and enteric nervous system. µ-opioid receptors (MOPs) are expressed and functional at the terminals of sensory axons, when activated by exogenous or endogenous ligands. However, the presence and function of MOP along nociceptive axons remains controversial particularly in naïve animals. Here, we characterized axonal MOPs by immunofluorescence, ultrastructural, and functional analyses. Furthermore, we evaluated hypertonic saline as a possible enhancer of opioid receptor function. RESULTS Comparative immunolabeling showed that, among several tested antibodies, which all provided specific MOP detection in the rat central nervous system (CNS), only one monoclonal MOP-antibody yielded specificity and reproducibility for MOP detection in the rat peripheral nervous system including the sciatic nerve. Double immunolabeling documented that MOP immunoreactivity was confined to calcitonin gene-related peptide (CGRP) positive fibers and fiber bundles. Almost identical labeling and double labeling patterns were found using mcherry-immunolabeling on sciatic nerves of mice producing a MOP-mcherry fusion protein (MOP-mcherry knock-in mice). Preembedding immunogold electron microscopy on MOP-mcherry knock-in sciatic nerves indicated presence of MOP in cytoplasm and at membranes of unmyelinated axons. Application of [D-Ala(2), N-MePhe(4), Gly-ol]-enkephalin (DAMGO) or fentanyl dose-dependently inhibited depolarization-induced CGRP release from rat sciatic nerve axons ex vivo, which was blocked by naloxone. When the lipophilic opioid fentanyl was applied perisciatically in naïve Wistar rats, mechanical nociceptive thresholds increased. Subthreshold doses of fentanyl or the hydrophilic opioid DAMGO were only effective if injected together with hypertonic saline. In vitro, using β-arrestin-2/MOP double-transfected human embryonic kidney cells, DAMGO as well as fentanyl lead to a recruitment of β-arrestin-2 to the membrane followed by a β-arrestin-2 reappearance in the cytosol and MOP internalization. Pretreatment with hypertonic saline prevented MOP internalization. CONCLUSION MOPs are present and functional in the axonal membrane from naïve animals. Hypertonic saline acutely decreases ligand-induced internalization of MOP and thereby might improve MOP function. Further studies should explore potential clinical applications of opioids together with enhancers for regional analgesia.
Collapse
Affiliation(s)
- Egle M Mambretti
- Department of Anesthesiology, University Hospital of Wuerzburg, Germany Institute of Anatomy and Cell Biology, University of Wuerzburg, Germany
| | - Katrin Kistner
- Institute of Physiology and Pathophysiology, University of Erlangen-Nuremberg, Germany
| | - Stefanie Mayer
- Institute for Pharmacology and Toxicology & Bio-Imaging Center/Rudolf-Virchow Center, University of Wuerzburg, Germany
| | - Dominique Massotte
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR, Strasbourg Cedex, France
| | - Brigitte L Kieffer
- Douglas Research Center, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Illkirch, France
| | - Carsten Hoffmann
- Institute for Pharmacology and Toxicology & Bio-Imaging Center/Rudolf-Virchow Center, University of Wuerzburg, Germany
| | - Peter W Reeh
- Institute of Physiology and Pathophysiology, University of Erlangen-Nuremberg, Germany
| | - Alexander Brack
- Department of Anesthesiology, University Hospital of Wuerzburg, Germany
| | - Esther Asan
- Institute of Anatomy and Cell Biology, University of Wuerzburg, Germany
| | - Heike L Rittner
- Department of Anesthesiology, University Hospital of Wuerzburg, Germany
| |
Collapse
|
33
|
“Barcode” and Differential Effects of GPCR Phosphorylation by Different GRKs. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3798-1_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
34
|
Moulédous L, Froment C, Burlet-Schiltz O, Schulz S, Mollereau C. Phosphoproteomic analysis of the mouse brain mu-opioid (MOP) receptor. FEBS Lett 2015; 589:2401-8. [PMID: 26226422 DOI: 10.1016/j.febslet.2015.07.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/03/2015] [Accepted: 07/20/2015] [Indexed: 11/19/2022]
Abstract
Many in vitro data have shown that the efficacy of several opioid drugs is correlated with differential mu-opioid (MOP) receptor phosphorylation. Label-free semiquantitative on-line nanoflow liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) analyses were performed to compare the endogenous MOP receptor phosphorylation patterns of mice administered with morphine, etonitazene and fentanyl. The analysis identified S363, T370 and S375 as phosphorylated residues in the carboxy-terminus. Only T370 and S375 were regulated by agonists, with a higher propensity to promote double phosphorylation for high efficacy agonists. Our study provides confirmation that differential agonist-driven multi-site phosphorylation of MOP receptor occurs in vivo and validate the use of MS to study endogenous GPCR phosphorylation.
Collapse
Affiliation(s)
- Lionel Moulédous
- Institut de Pharmacologie et Biologie Structurale CNRS/Université de Toulouse, 205 route de Narbonne, 31077 Toulouse, France
| | - Carine Froment
- Institut de Pharmacologie et Biologie Structurale CNRS/Université de Toulouse, 205 route de Narbonne, 31077 Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et Biologie Structurale CNRS/Université de Toulouse, 205 route de Narbonne, 31077 Toulouse, France
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Strasse 1, 07747 Jena, Germany.
| | - Catherine Mollereau
- Institut de Pharmacologie et Biologie Structurale CNRS/Université de Toulouse, 205 route de Narbonne, 31077 Toulouse, France.
| |
Collapse
|
35
|
Habibi-Asl B, Vaez H, Najafi M, Bidaghi A, Ghanbarzadeh S. Attenuation of morphine-induced dependence and tolerance by ceftriaxone and amitriptyline in mice. ACTA ACUST UNITED AC 2014; 52:163-8. [PMID: 25557842 DOI: 10.1016/j.aat.2014.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 11/07/2014] [Accepted: 11/12/2014] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Tolerance to and dependence on the analgesic effect of opioids is a pharmacological phenomenon that occurs after their prolonged administration. OBJECTIVE The aim of this study was to evaluate the protective effects of ceftriaxone and amitriptyline on the development of morphine-induced tolerance and dependence. METHODS In this study, 18 groups (9 groups each for tolerance and dependency tests) of mice (n = 8) received saline [10 mL/kg, intraperitoneally (i.p.)], morphine (50 mg/kg, i.p.), ceftriaxone (50 mg/kg, i.p., 100 mg/kg, i.p., and 200 mg/kg, i.p.), amitriptyline (5 mg/kg, i.p., 10 mg/kg, i.p., and 15 mg/kg, i.p.), or a combination of ceftriaxone (50 mg/kg, i.p.) and amitriptyline (5 mg/kg, i.p.) once per day for 4 days for investigation and comparison of the effects of ceftriaxone and amitriptyline on the prevention of dependency and tolerance to morphine. Tolerance was assessed with administration of morphine (9 mg/kg, i.p.) and using the hot plate test on the 5(th) day. In dependency tests, withdrawal symptoms were assessed on the 4(th) day for each animal 30 minutes after the administration of naloxone (4 mg/kg, i.p.; 2 hours after the last dose of morphine). RESULTS It was found that treatment with ceftriaxone or amitriptyline attenuated the development of tolerance to the antinociceptive effect of morphine and also reduced naloxone-precipitated withdrawal jumping and standing on feet. Furthermore, coadministration of ceftriaxone and amitriptyline at low doses (50 mg/kg, i.p. and 5 mg/kg, i.p., respectively) prior to morphine injection also decreased both morphine-induced tolerance and dependence. CONCLUSION Results indicate that the treatment with ceftriaxone and amitriptyline, alone or in combination, could attenuate the development of morphine-induced tolerance and dependence.
Collapse
Affiliation(s)
- Bohlul Habibi-Asl
- Department of Pharmacology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haleh Vaez
- Department of Pharmacology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moslem Najafi
- Department of Pharmacology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Bidaghi
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Ghanbarzadeh
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
36
|
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
|
37
|
Bray L, Froment C, Pardo P, Candotto C, Burlet-Schiltz O, Zajac JM, Mollereau C, Moulédous L. Identification and functional characterization of the phosphorylation sites of the neuropeptide FF2 receptor. J Biol Chem 2014; 289:33754-66. [PMID: 25326382 DOI: 10.1074/jbc.m114.612614] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The neuropeptide FF2 (NPFF2) receptor belongs to the rhodopsin family of G protein-coupled receptors and mediates the effects of several related RFamide neuropeptides. One of the main pharmacological interests of this system resides in its ability to regulate endogenous opioid systems, making it a potential target to reduce the negative effects of chronic opioid use. Phosphorylation of intracellular residues is the most extensively studied post-translational modification regulating G protein-coupled receptor activity. However, until now, no information concerning NPFF2 receptor phosphorylation is available. In this study, we combined mass spectrometric analysis and site-directed mutagenesis to analyze for the first time the phosphorylation pattern of the NPFF2 receptor and the role of the various phosphorylation sites in receptor signaling, desensitization, and trafficking in a SH-SY5Y model cell line. We identified the major, likely GRK-dependent, phosphorylation cluster responsible for acute desensitization, (412)TNST(415) at the end of the C terminus of the receptor, and additional sites involved in desensitization ((372)TS(373)) and internalization (Ser(395)). We thus demonstrate the key role played by phosphorylation in the regulation of NPFF2 receptor activity and trafficking. Our data also provide additional evidence supporting the concept that desensitization and internalization are partially independent processes relying on distinct phosphorylation patterns.
Collapse
Affiliation(s)
- Lauriane Bray
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Carine Froment
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Pierre Pardo
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Cédric Candotto
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Odile Burlet-Schiltz
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Jean-Marie Zajac
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Catherine Mollereau
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Lionel Moulédous
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| |
Collapse
|