دورية أكاديمية

A novel G protein-biased agonist at the μ opioid receptor induces substantial receptor desensitisation through G protein-coupled receptor kinase.

التفاصيل البيبلوغرافية
العنوان: A novel G protein-biased agonist at the μ opioid receptor induces substantial receptor desensitisation through G protein-coupled receptor kinase.
المؤلفون: Groom S; Department of Pharmacy and Pharmacology, University of Bath, Bath, UK.; School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK., Blum NK; Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany., Conibear AE; School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK., Disney A; Department of Pharmacy and Pharmacology, University of Bath, Bath, UK., Hill R; School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK.; School of Life Sciences, University of Nottingham, Nottingham, UK., Husbands SM; Department of Pharmacy and Pharmacology, University of Bath, Bath, UK., Li Y; Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, South Carolina, USA., Toll L; Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA., Kliewer A; Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany., Schulz S; Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany., Henderson G; School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK., Kelly E; School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK., Bailey CP; Department of Pharmacy and Pharmacology, University of Bath, Bath, UK.
المصدر: British journal of pharmacology [Br J Pharmacol] 2023 Apr; Vol. 180 (7), pp. 943-957. Date of Electronic Publication: 2020 Dec 27.
نوع المنشور: Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov't
اللغة: English
بيانات الدورية: Publisher: Wiley Country of Publication: England NLM ID: 7502536 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1476-5381 (Electronic) Linking ISSN: 00071188 NLM ISO Abbreviation: Br J Pharmacol Subsets: MEDLINE
أسماء مطبوعة: Publication: London : Wiley
Original Publication: London, Macmillian Journals Ltd.
مواضيع طبية MeSH: Analgesics, Opioid*/pharmacology , Receptors, Opioid, mu*/metabolism, Rats ; Humans ; Animals ; GTP-Binding Proteins/metabolism ; Morphine/pharmacology ; Signal Transduction ; G-Protein-Coupled Receptor Kinases/metabolism ; Mammals/metabolism
مستخلص: Background and Purpose: G protein-biased μ opioid receptor agonists have the potential to induce less receptor desensitisation and tolerance than balanced opioids. Here, we investigated if the cyclic endomorphin analogue Tyr-c[D-Lys-Phe-Tyr-Gly] (Compound 1) is a G protein-biased μ agonist and characterised its ability to induce rapid receptor desensitisation in mammalian neurones.
Experimental Approach: The signalling and trafficking properties of opioids were characterised using bioluminescence resonance energy transfer assays, enzyme-linked immunosorbent assay and phosphosite-specific immunoblotting in human embryonic kidney 293 cells. Desensitisation of opioid-induced currents were studied in rat locus coeruleus neurones using whole-cell patch-clamp electrophysiology. The mechanism of Compound 1-induced μ receptor desensitisation was probed using kinase inhibitors.
Key Results: Compound 1 has similar intrinsic activity for G protein signalling as morphine. As predicted for a G protein-biased μ agonist, Compound 1 induced minimal agonist-induced internalisation and phosphorylation at intracellular μ receptor serine/threonine residues known to be involved in G protein-coupled receptor kinase (GRK)-mediated desensitisation. However, Compound 1 induced robust rapid μ receptor desensitisation in locus coeruleus neurons, to a greater degree than morphine. The extent of Compound 1-induced desensitisation was unaffected by activation or inhibition of protein kinase C (PKC) but was significantly reduced by inhibition of GRK.
Conclusion and Implications: Compound 1 is a novel G protein-biased μ agonist that induces substantial rapid receptor desensitisation in mammalian neurons. Surprisingly, Compound 1-induced desensitisation was demonstrated to be GRK dependent despite its G protein bias. Our findings refute the assumption that G protein-biased agonists will evade receptor desensitisation and tolerance.
Linked Articles: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
(© 2020 The British Pharmacological Society.)
References: Alexander, S. P. H., Christopoulos, A., Davenport, A. P., Kelly, E., Mathie, A., Peters, J. A., … CGTP Collaborators. (2019). THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: G protein-coupled receptors. British Journal of Pharmacology, 176(S1), S21-S141.
Alexander, S. P. H., Fabbro, D., Kelly, E., Mathie, A., Peters, J. A., Veale, E. L., … CGTP Collaborators. (2019). THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: Enzymes. British Journal of Pharmacology, 176(S1), S297-S396.
Alexander, S. P. H., Mathie, A., Peters, J. A., Veale, E. L., Striessnig, J., Kelly, E., … CGTP Collaborators. (2019). THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: Ion channels. British Journal of Pharmacology, 176(S1), S142-S228.
Alexander, S. P. H., Roberts, R. E., Broughton, B. R. S., Sobey, C. G., George, C. H., Stanford, S. C., … Ahluwalia, A. (2018). Goals and practicalities of immunoblotting and immunohistochemistry: A guide for submission to the British Journal of Pharmacology. British Journal of Pharmacology, 175, 407-411. https://doi.org/10.1111/bph.14112.
Altarifi, A. A., David, B., Muchhala, K. H., Blough, B. E., Akbarali, H., & Negus, S. S. (2017). Effects of acute and repeated treatment with the biased μ opioid receptor agonist TRV130 (oliceridine) on measures of antinociception, gastrointestinal function, and abuse liability in rodents. Journal of Psychopharmacology, 31(6), 730-739. https://doi.org/10.1177/0269881116689257.
Bailey, C. P., Kelly, E., & Henderson, G. (2004). Protein kinase C activation enhances morphine-induced rapid desensitization of μ opioid receptors in mature rat locus ceruleus neurons. Molecular Pharmacology, 66(6), 1592-1598. https://doi.org/10.1124/mol.104.004747.
Bailey, C. P., Llorente, J., Gabra, B. H., Smith, F. L., Dewey, W. L., Kelly, E., & Henderson, G. (2009). Role of protein kinase C and μ opioid receptor (MOPr) desensitization in tolerance to morphine in rat locus coeruleus neurons. The European Journal of Neuroscience, 29(2), 307-318. https://doi.org/10.1111/j.1460-9568.2008.06573.x.
Bailey, C. P., Oldfield, S., Llorente, J., Caunt, C. J., Teschemacher, A. G., Roberts, L., … Henderson, G. (2009). Involvement of PKC alpha and G-protein-coupled receptor kinase 2 in agonist-selective desensitization of μ opioid receptors in mature brain neurons. British Journal of Pharmacology, 158(1), 157-164. https://doi.org/10.1111/j.1476-5381.2009.00140.x.
Bohn, L. M., Gainetdinov, R. R., Lin, F. T., Lefkowitz, R. J., & Caron, M. G. (2000). μ opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature, 408(6813), 720-723. https://doi.org/10.1038/35047086.
Bohn, L. M., Lefkowitz, R. J., Gainetdinov, R. R., Peppel, K., Caron, M. G., & Lin, F. T. (1999). Enhanced morphine analgesia in mice lacking beta-arrestin 2. Science, 286(5449), 2495-2498. https://doi.org/10.1126/science.286.5449.2495.
Celver, J., Xu, M., Jin, W., Lowe, J., & Chavkin, C. (2004). Distinct domains of the μ opioid receptor control uncoupling and internalization. Molecular Pharmacology, 65(3), 528-537. https://doi.org/10.1124/mol.65.3.528.
Celver, J. P., Lowe, J., Kovoor, A., Gurevich, V. V., & Chavkin, C. (2001). Threonine 180 is required for G-protein-coupled receptor kinase 3- and beta-arrestin 2-mediated desensitization of the μ opioid receptor in Xenopus oocytes. The Journal of Biological Chemistry, 276(7), 4894-4900. https://doi.org/10.1074/jbc.M007437200.
Cheng, Z. J., Yu, Q. M., Wu, Y. L., Ma, L., & Pei, G. (1998). Selective interference of beta-arrestin 1 with kappa and delta but not μ opioid receptor/G protein coupling. The Journal of Biological Chemistry, 273(38), 24328-24333. https://doi.org/10.1074/jbc.273.38.24328.
Clayton, C. C., Donthamsetti, P., Lambert, N. A., Javitch, J. A., & Neve, K. A. (2014). Mutation of three residues in the third intracellular loop of the dopamine D2 receptor creates an internalization-defective receptor. The Journal of Biological Chemistry, 289(48), 33663-33675. https://doi.org/10.1074/jbc.M114.605378.
Conibear, A. E., & Kelly, E. (2019). A biased view of μ opioid receptors? Molecular Pharmacology, 96(5), 542-549. https://doi.org/10.1124/mol.119.115956.
Curtis, M. J., Alexander, S., Cirino, G., Docherty, J. R., George, C. H., Giembycz, M. A., … Ahluwalia, A. (2018). Experimental design and analysis and their reporting II: Updated and simplified guidance for authors and peer reviewers. British Journal of Pharmacology, 175(7), 987-993. https://doi.org/10.1111/bph.14153.
Dekan, Z., Sianati, S., Yousuf, A., Sutcliffe, K. J., Gillis, A., Mallet, C., … Christie, M. J. (2019). A tetrapeptide class of biased analgesics from an Australian fungus targets the μ opioid receptor. Proceedings of the National Academy of Sciences of the United States of America, 116(44), 22353-22358. https://doi.org/10.1073/pnas.1908662116.
DeWire, S. M., Yamashita, D. S., Rominger, D. H., Liu, G., Cowan, C. L., Graczyk, T. M., … Violin, J. D. (2013). A G protein-biased ligand at the μ opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphine. The Journal of Pharmacology and Experimental Therapeutics, 344(3), 708-717. https://doi.org/10.1124/jpet.112.201616.
Dhami, G. K., Babwah, A. V., Sterne-Marr, R., & Ferguson, S. S. (2005). Phosphorylation-independent regulation of metabotropic glutamate receptor 1 signaling requires G protein-coupled receptor kinase 2 binding to the second intracellular loop. The Journal of Biological Chemistry, 280(26), 24420-24427. https://doi.org/10.1074/jbc.M501650200.
Gillis, A., Gondin, A. B., Kliewer, A., Sanchez, J., Lim, H. D., Alamein, C., … Canals, M. (2020). Low intrinsic efficacy for G protein activation can explain the improved side effect profiles of new opioid agonists. Science Signaling, 13(625), eaaz3140. https://doi.org/10.1016/j.jss.2020.09.026.
Grim, T. W., Schmid, C. L., Stahl, E. L., Pantouli, F., Ho, J. H., Acevedo-Canabal, A., … Bohn, L. M. (2020). A G protein signaling-biased agonist at the μ opioid receptor reverses morphine tolerance while preventing morphine withdrawal. Neuropsychopharmacology, 45(2), 416-425. https://doi.org/10.1038/s41386-019-0491-8.
Harris, G. C., & Williams, J. T. (1991). Transient homologous mu-opioid receptor desensitization in rat locus coeruleus neurons. Journal of Neuroscience, 11(9), 2574-2681.
Hill, R., Dewey, W. L., Kelly, E., & Henderson, G. (2018). Oxycodone-induced tolerance to respiratory depression: Reversal by ethanol, pregabalin and protein kinase C inhibition. British Journal of Pharmacology, 175(12), 2492-2503. https://doi.org/10.1111/bph.14219.
Hill, R., Disney, A., Conibear, A., Sutcliffe, K., Dewey, W., Husbands, S., … Henderson, G. (2018). The novel μ opioid receptor agonist PZM21 depresses respiration and induces tolerance to antinociception. British Journal of Pharmacology, 175(13), 2653-2661. https://doi.org/10.1111/bph.14224.
Hull, L. C., Llorente, J., Gabra, B. H., Smith, F. L., Kelly, E., Bailey, C., … Dewey, W. L. (2010). The effect of protein kinase C and G protein-coupled receptor kinase inhibition on tolerance induced by μ opioid agonists of different efficacy. The Journal of Pharmacology and Experimental Therapeutics, 332(3), 1127-1135. https://doi.org/10.1124/jpet.109.161455.
Johnson, E. A., Oldfield, S., Braksator, E., Gonzalez-Cuello, A., Couch, D., Hall, K. J., … Henderson, G. (2006). Agonist-selective mechanisms of μ opioid receptor desensitization in human embryonic kidney 293 cells. Molecular Pharmacology, 70(2), 676-685. https://doi.org/10.1124/mol.106.022376.
Just, S., Illing, S., Trester-Zedlitz, M., Lau, E. K., Kotowski, S. J., Miess, E., … Schulz, S. (2013). Differentiation of opioid drug effects by hierarchical multi-site phosphorylation. Molecular Pharmacology, 83(3), 633-639. https://doi.org/10.1124/mol.112.082875.
Kelly, E. (2013). Efficacy and ligand bias at the mu-opioid receptor. British Journal of Pharmacology, 169(7), 1430-1446. https://doi.org/10.1111/bph.12222.
Kelly, E., Bailey, C. P., & Henderson, G. (2008). Agonist-selective mechanisms of GPCR desensitization. British Journal of Pharmacology, 153(S1), S379-S388.
Kenakin, T. (2019). Biased receptor signaling in drug discovery. Pharmacological Reviews, 71(2), 267-315. https://doi.org/10.1124/pr.118.016790.
Kilkenny, C., Browne, W. J., Cuthill, I. C., Emerson, M., & Altman, D. G. (2010). Improving bioscience research reporting: The ARRIVE guidelines for reporting animal research. PLoS Biology, 8(6), e1000412. https://doi.org/10.1371/journal.pbio.1000412.
Kliewer, A., Gillis, A., Hill, R., Schmidel, F., Bailey, C., Kelly, E., … Schulz, S. (2020). Morphine-induced respiratory depression is independent of beta-arrestin 2 signalling. British Journal of Pharmacology, 177, 2923-2931. https://doi.org/10.1111/bph.15004.
Kliewer, A., Schmiedel, F., Sianati, S., Bailey, A., Bateman, J. T., Levitt, E. S., … Schulz, S. (2019). Phosphorylation-deficient G-protein-biased μ opioid receptors improve analgesia and diminish tolerance but worsen opioid side effects. Nature Communications, 10(1), 367. https://doi.org/10.1038/s41467-018-08162-1.
Li, Y., Cazares, M., Wu, J., Houghten, R. A., Toll, L., & Dooley, C. (2016). Potent μ opioid receptor agonists from cyclic peptides Tyr-c[D-Lys-xxx-Tyr-Gly]: Synthesis, biological, and structural evaluation. Journal of Medicinal Chemistry, 59(3), 1239-1245. https://doi.org/10.1021/acs.jmedchem.5b01899.
Liang, D.-Y., Li, W.-W., Nwaneshiudu, C., Irvine, K.-A., & Clark, J. D. (2019). Pharmacological characters of oliceridine, a μ-opioid receptor G-protein-biased ligand in mice. Anesthesia & Analgesia, 129(5), 1414-1421. https://doi.org/10.1213/ANE.0000000000003662.
Llorente, J., Lowe, J. D., Sanderson, H. S., Tsisanova, E., Kelly, E., Henderson, G., & Bailey, C. P. (2012). μ opioid receptor desensitization: Homologous or heterologous? The European Journal of Neuroscience, 36(12), 3636-3642. https://doi.org/10.1111/ejn.12003.
Lowe, J. D., Sanderson, H. S., Cooke, A. E., Ostovar, M., Tsisanova, E., Withey, S. L., … Bailey, C. P. (2015). Role of G protein-coupled receptor kinases 2 and 3 in μ opioid receptor desensitization and internalization. Molecular Pharmacology, 88(2), 347-356. https://doi.org/10.1124/mol.115.098293.
Manglik, A., Lin, H., Aryal, D. K., McCorvy, J. D., Dengler, D., Corder, G., … Shoichet, B. K. (2016). Structure-based discovery of opioid analgesics with reduced side effects. Nature, 537(7619), 185-190. https://doi.org/10.1038/nature19112.
Matthes, H. W., Maldonado, R., Simonin, F., Valverde, O., Slowe, S., Kitchen, I., … Kieffer, B. L. (1996). Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the μ opioid-receptor gene. Nature, 383(6603), 819-823. https://doi.org/10.1038/383819a0.
McFadzean, I., Lacey, M. G., Hill, R. G., & Henderson, G. (1987). Kappa opioid receptor activation depresses excitatory synaptic input to rat locus coeruleus neurons in vitro. Neuroscience, 20(1), 231-239. https://doi.org/10.1016/0306-4522(87)90015-7.
Miess, E., Gondin, A. B., Yousuf, A., Steinborn, R., Mosslein, N., Yang, Y., … Canals, M. (2018). Multisite phosphorylation is required for sustained interaction with GRKs and arrestins during rapid μ opioid receptor desensitization. Science Signaling, 11(539), eaas9609. https://doi.org/10.1126/scisignal.aas9609.
North, R. A., & Williams, J. T. (1985). On the potassium conductance increased by opioids in rat locus coeruleus neurones. The Journal of Physiology, 364(1), 265-280. https://doi.org/10.1113/jphysiol.1985.sp015743.
Pals-Rylaarsdam, R., & Hosey, M. M. (1997). Two homologous phosphorylation domains differentially contribute to desensitization and internalization of the M2 muscarinic acetylcholine receptor. The Journal of Biological Chemistry, 272(22), 14152-14158. https://doi.org/10.1074/jbc.272.22.14152.
Raehal, K. M., Walker, J. K., & Bohn, L. M. (2005). Morphine side effects in beta-arrestin 2 knockout mice. The Journal of Pharmacology and Experimental Therapeutics, 314(3), 1195-1201. https://doi.org/10.1124/jpet.105.087254.
Raveh, A., Cooper, A., Guy-David, L., & Reuveny, E. (2010). Nonenzymatic rapid control of GIRK channel function by a G protein-coupled receptor kinase. Cell, 143(5), 750-760. https://doi.org/10.1016/j.cell.2010.10.018.
Ribeiro, F. M., Ferreira, L. T., Paquet, M., Cregan, T., Ding, Q., Gros, R., & Ferguson, S. S. (2009). Phosphorylation-independent regulation of metabotropic glutamate receptor 5 desensitization and internalization by G protein-coupled receptor kinase 2 in neurons. The Journal of Biological Chemistry, 284(35), 23444-23453. https://doi.org/10.1074/jbc.M109.000778.
Rivero, G., Llorente, J., McPherson, J., Cooke, A., Mundell, S. J., McArdle, C. A., … Kelly, E. (2012). Endomorphin-2: A biased agonist at the μ opioid receptor. Molecular Pharmacology, 82(2), 178-188. https://doi.org/10.1124/mol.112.078659.
Rodriguez-Martin, I., Braksator, E., Bailey, C. P., Goodchild, S., Marrion, N. V., Kelly, E., & Henderson, G. (2008). Methadone: Does it really have low efficacy at μ opioid receptors? Neuroreport, 19(5), 589-593. https://doi.org/10.1097/WNR.0b013e3282f97b64.
Schmid, C. L., Kennedy, N. M., Ross, N. C., Lovell, K. M., Yue, Z., Morgenweck, J., … Bohn, L. M. (2017). Bias factor and therapeutic window correlate to predict safer opioid analgesics. Cell, 171(5), 1165-1175, e1113. https://doi.org/10.1016/j.cell.2017.10.035.
Schneider, S., Provasi, D., & Filizola, M. (2016). How oliceridine (TRV-130) binds and stabilizes a μ opioid receptor conformational state that selectively triggers G protein signaling pathways. Biochemistry, 55(46), 6456-6466. https://doi.org/10.1021/acs.biochem.6b00948.
van Bockstaele, E. J., Commons, K., & Pickel, V. M. (1997). Delta-opioid receptor is present in presynaptic axon terminals in the rat nucleus locus coeruleus: Relationships with methionine 5-enkephalin. The Journal of Comparative Neurology, 388(4), 575-586. https://doi.org/10.1002/(SICI)1096-9861(19971201)388:4<575::AID-CNE6>3.0.CO;2-#.
Williams, J. T., Ingram, S. L., Henderson, G., Chavkin, C., von Zastrow, M., Schulz, S., … Christie, M. J. (2013). Regulation of μ opioid receptors: Desensitization, phosphorylation, internalization, and tolerance. Pharmacological Reviews, 65(1), 223-254. https://doi.org/10.1124/pr.112.005942.
Withey, S. L., Hill, R., Lyndon, A., Dewey, W. L., Kelly, E., & Henderson, G. (2017). Effect of tamoxifen and brain-penetrant protein kinase C and c-Jun N-terminal kinase inhibitors on tolerance to opioid-induced respiratory depression in mice. Journal of Pharmacology and Experimental Therapeutics, 361(1), 51-59. https://doi.org/10.1124/jpet.116.238329.
Yudin, Y., & Rohacs, T. (2019). The G-protein-biased agents PZM21 and TRV130 are partial agonists of mu-opioid receptor-mediated signalling to ion channels. British Journal of Pharmacology, 176(17), 3110-3125. https://doi.org/10.1111/bph.14702.
Zhang, J., Ferguson, S. S., Barak, L. S., Bodduluri, S. R., Laporte, S. A., Law, P. Y., & Caron, M. G. (1998). Role for G protein-coupled receptor kinase in agonist-specific regulation of μ opioid receptor responsiveness. Proceedings of the National Academy of Sciences of the United States of America, 95(12), 7157-7162. https://doi.org/10.1073/pnas.95.12.7157.
معلومات مُعتمدة: British Pharmacological Society (BPS); R01 DA007315 United States DA NIDA NIH HHS; R01 DA036975 United States DA NIDA NIH HHS
فهرسة مساهمة: Keywords: G protein-coupled receptor kinases; arrestins; biased agonism; electrophysiology; opiates; opioids; receptor desensitisation
المشرفين على المادة: 0 (Analgesics, Opioid)
0 (Receptors, Opioid, mu)
EC 3.6.1.- (GTP-Binding Proteins)
76I7G6D29C (Morphine)
EC 2.7.11.16 (G-Protein-Coupled Receptor Kinases)
تواريخ الأحداث: Date Created: 20201127 Date Completed: 20230303 Latest Revision: 20230314
رمز التحديث: 20240628
DOI: 10.1111/bph.15334
PMID: 33245558
قاعدة البيانات: MEDLINE
الوصف
تدمد:1476-5381
DOI:10.1111/bph.15334