Bias-Inducing Allosteric Binding Site in Mu-Opioid Receptor Signaling

Total Page:16

File Type:pdf, Size:1020Kb

Bias-Inducing Allosteric Binding Site in Mu-Opioid Receptor Signaling Research Article Bias‑inducing allosteric binding site in mu‑opioid receptor signaling Andrés F. Marmolejo‑Valencia1 · Abraham Madariaga‑Mazón1 · Karina Martinez‑Mayorga1 Received: 23 January 2021 / Accepted: 12 March 2021 © The Author(s) 2021 OPEN Abstract G-protein-biased agonism of the mu-opioid receptor (μ-OR) is emerging as a promising strategy in analgesia. A deep understanding of how biased agonists modulate and diferentiate G-protein-coupled receptors (GPCR) signaling path- ways and how this is transferred into the cell are open questions. Here, using extensive all-atom molecular dynamics simulations, we analyzed the binding recognition process and signaling efects of three prototype μ-OR agonists. Our suggested structural mechanism of biased signaling in μ-OR involves an allosteric sodium ion site, water networks, confor- mational rearrangements in conserved motifs and collective motions of loops and transmembrane helices. These analyses led us to highlight the relevance of a bias-inducing allosteric binding site in the understanding of μ-OR’s G-protein-biased signaling. These results also suggest a competitive equilibrium between the agonists and the allosteric sodium ion, where the bias-inducing allosteric binding site can be modulated by this ion or an agonist such as herkinorin. Notably, herkinorin arises as the archetype modulator of μ-OR and its interactive pattern could be used for screening eforts via protein–ligand interaction fngerprint (PLIF) studies. Article highlights • Agonists and a sodium ion compete for the bias-inducing allosteric binding site that modulates signaling in mu-opioid receptors. • Molecular dynamics simulations of the prototype μ-OR agonist suggest a competitive equilibrium involving the agonist and an allosteric sodium ion. • Analysis of experimental data from the literature and molecular models provides the structural bases of biased ago- nism on μ-OR. Keywords Biased signaling · GPCR · Mu-opioid receptor · Molecular dynamics simulations 1 Introduction G-proteins and/or with β-arrestins. Preferential activation to either pathway is possible, leading to diferent physi- G-protein-coupled receptors (GPCRs) are seven-trans- ological efects. Agonists that have this ability are called membrane (TM) helical proteins that trigger signaling biased agonists, and the efect is named functional selec- pathways, mediated by interactions with heterotrimeric tivity. Other modes of GPCR activation are intracellular Supplementary Information The online version contains supplementary material available at https:// doi. org/ 10. 1007/ s42452- 021- 04505-8. * Karina Martinez-Mayorga, [email protected] | 1Instituto de Química, Universidad Nacional Autónoma de México, Mexico City, México. SN Applied Sciences (2021) 3:566 | https://doi.org/10.1007/s42452-021-04505-8 Vol.:(0123456789) Research Article SN Applied Sciences (2021) 3:566 | https://doi.org/10.1007/s42452-021-04505-8 activation, dimerization activation, transactivation, or relevance of TM7 movement is suggested in arginine- biphasic activation [1]. vasopressin type 2 receptor (V2R) [14] using fuorescence Biased activation of the mu-opioid receptor (μ-OR) spectroscopy; and in β2-adrenergic receptors (β2AR) [15] towards G-protein is relevant for the design of painkillers characterized by 19F-NMR experiments. These studies with reduced detrimental side efects. For example, activa- revealed that movements at TM6 helix are required for tion of β-arrestin signaling pathways is suggested to lead G-protein signaling. In contrast, β-arrestin-biased ligands to tolerance, desensitization, addiction, constipation, and impact the TM7-H8 region. This information could be respiratory suppression [2]. Albeit not fully understood, transferable across class A GPCRs [7, 8, 16, 17] where the these efects seem to be related to β-arrestin recruitment allosteric sodium site is preserved. To note, experimental or receptor internalization. Moreover, the coexistence of information regarding the allosteric sodium site in μ-OR diverse and complex GPCR activation types is an active is scarce [18, 19], but many computational studies have research topic. This complexity is further evidenced by the shown the collapse of the allosteric sodium site in the ability of GPCRs to adopt diferent conformations. Experi- activation and the egress pathways of the sodium ion in mental evidence shows that diferent agonists can stabi- the presence of agonists and modulators [19–21]. These lize these conformations, leading to new conformational studies also suggest the lack of spontaneous release of landscapes [3–5]. This behavior is counterintuitive, based the sodium ion from the bundle which required random on the paradigm that all agonists would conduct to only accelerated molecular dynamics simulations to explore one active conformation [6]. possible outlets from the allosteric site [19]. Besides the biased ligands, experimental evidence This important behavior has motivated the search shows that a conserved allosteric binding site plays a for allosteric modulators in μ-OR. For example, ignavine crucial role in the activation of class A GPCRs that include increases agonist response for DAMGO and has positive μ-OR [7]. Modulation of the GPCRs via this allosteric site is modulatory activity in morphine and DAMGO assays. Fur- related to downstream signaling and physiological efects thermore, ignavine produces an analgesic efect in vivo [8, 9]. The high-resolution (1.8 Å) X-ray crystal structure through opioid response, proved by tail-flick and tail of the adenosine A2A receptor revealed a highly ordered pressure tests [22]. However, the mechanistic modula- cluster of atoms, composed of one sodium ion, water mol- tion transmission in opioid receptors is still unclear. The ecules, and conserved amino acids [8] located on TM2 and positive allosteric modulator (PAM) BMS-986122 of μ-OR, TM3, such as D 2.50 and S 3.39. The importance of an allos- which suppresses the β-arrestin recruitment in conjunc- teric sodium binding site nearby D2.50 has been reviewed tion with agonists [23], can decrease the ability of NaCl to [7–9], particularly on the receptor activation process and modulate the binding afnity of agonists and has been G-protein/arrestin signaling bias. Also, the structure of the proposed to act as an indirect competitor of the allosteric delta-opioid receptor (δ-OR), in the inactive state, showed sodium ion [18]. Additionally, molecular dynamics (MD) an electron density distribution located in an allosteric site simulations of BMS-986122 suggested that residues on in the vicinity of D2.50, corresponding to a sodium ion and TM7 (7.53 and 7.35) are involved in the positive allos- also unveiled N3.35 as an important amino acid in this bind- teric signal transmission [24]. Notably, the structure of ing site [9]. In agreement with these fndings, mutations BMS-986122 contains motifs related to C2-benzyloxy of N3.35 V and N 3.35A decreased and abolished δ-OR agonist herkinorin; this is in agreement with our suggested bind- sodium efect, respectively. Interestingly, in both cases, the ing mode of herkinorin involving modulation of TM7 [25]. δ-OR arrestin-mediated signaling pathway is increased. Comparing the X-ray structures of μ-OR in the active However, both mutations did not afect the canonical sign- [26] and inactive [27] states allows analyzing helical rear- aling, which is mediated by G-proteins. This suggests that rangements related to the activation process. These rear- while the allosteric sodium ion can modulate the recruit- rangements are common to other members of the class ment of β-arrestin, the G-protein–receptor binding only A GPCR superfamily (β-2AR, rhodopsin, M2R) [7, 8, 16], depends on the agonist [9]. Initial experiments showed such as helical movements at TM6 and TM7. Moreover, a dependence on sodium concentration for the afnity changes in water-mediated interactions involving amino of agonists but not for antagonists [10, 11]. Antagonists/ acids inside the receptor’s cavity are observed when the weak partial agonists are also afected by D2.50 mutations, activation is established [26]. The active state of the recep- which become β-arrestin-biased agonist [7]. Other small tor is characterized by an ordered network involving water molecules such as amiloride can compete with the allos- molecules. Notably, careful analysis of the water network teric sodium site [9, 12, 13]. shows that it is functionally relevant, transferring infor- Mutations at D 2.50 and N 3.35 in δ-OR at varied concen- mation from the extracellular side (ligand binding site) tration of Na+ afect recruitment of β-arrestin, most likely through the receptor [26], and reaching the interior of the by affecting the TM7 helical bundle [9]. Similarly, the cell. This last step is characterized by collective movements Vol:.(1234567890) SN Applied Sciences (2021) 3:566 | https://doi.org/10.1007/s42452-021-04505-8 Research Article of intracellular regions of the TMs and loops. This informa- 2 Results tion relates to the exquisite modulation of the receptor that results from ligand binding. The μ-OR biased signaling mechanism, proposed in this Interestingly, herkinorin, a non-nitrogenous μ-OR work, initiated at the orthosteric binding site continues agonist, does not produce β-arrestin2 recruitment and via an allosteric binding site [26], then reaches a hydropho- μ-OR cell internalization, even when the G-protein- bic barrier (framed by DRY and NPxxY conserved motifs) coupled receptor kinase 2 (GRK2) is overexpressed. [34,
Recommended publications
  • INVESTIGATION of NATURAL PRODUCT SCAFFOLDS for the DEVELOPMENT of OPIOID RECEPTOR LIGANDS by Katherine M
    INVESTIGATION OF NATURAL PRODUCT SCAFFOLDS FOR THE DEVELOPMENT OF OPIOID RECEPTOR LIGANDS By Katherine M. Prevatt-Smith Submitted to the graduate degree program in Medicinal Chemistry and the Graduate Faculty of the University of Kansas in partial fulfillment of the requirements for the degree of Doctor of Philosophy. _________________________________ Chairperson: Dr. Thomas E. Prisinzano _________________________________ Dr. Brian S. J. Blagg _________________________________ Dr. Michael F. Rafferty _________________________________ Dr. Paul R. Hanson _________________________________ Dr. Susan M. Lunte Date Defended: July 18, 2012 The Dissertation Committee for Katherine M. Prevatt-Smith certifies that this is the approved version of the following dissertation: INVESTIGATION OF NATURAL PRODUCT SCAFFOLDS FOR THE DEVELOPMENT OF OPIOID RECEPTOR LIGANDS _________________________________ Chairperson: Dr. Thomas E. Prisinzano Date approved: July 18, 2012 ii ABSTRACT Kappa opioid (KOP) receptors have been suggested as an alternative target to the mu opioid (MOP) receptor for the treatment of pain because KOP activation is associated with fewer negative side-effects (respiratory depression, constipation, tolerance, and dependence). The KOP receptor has also been implicated in several abuse-related effects in the central nervous system (CNS). KOP ligands have been investigated as pharmacotherapies for drug abuse; KOP agonists have been shown to modulate dopamine concentrations in the CNS as well as attenuate the self-administration of cocaine in a variety of species, and KOP antagonists have potential in the treatment of relapse. One drawback of current opioid ligand investigation is that many compounds are based on the morphine scaffold and thus have similar properties, both positive and negative, to the parent molecule. Thus there is increasing need to discover new chemical scaffolds with opioid receptor activity.
    [Show full text]
  • Current Awareness in Clinical Toxicology Editors: Damian Ballam Msc and Allister Vale MD
    Current Awareness in Clinical Toxicology Editors: Damian Ballam MSc and Allister Vale MD February 2016 CONTENTS General Toxicology 9 Metals 38 Management 21 Pesticides 41 Drugs 23 Chemical Warfare 42 Chemical Incidents & 32 Plants 43 Pollution Chemicals 33 Animals 43 CURRENT AWARENESS PAPERS OF THE MONTH How toxic is ibogaine? Litjens RPW, Brunt TM. Clin Toxicol 2016; online early: doi: 10.3109/15563650.2016.1138226: Context Ibogaine is a psychoactive indole alkaloid found in the African rainforest shrub Tabernanthe Iboga. It is unlicensed but used in the treatment of drug and alcohol addiction. However, reports of ibogaine's toxicity are cause for concern. Objectives To review ibogaine's pharmacokinetics and pharmacodynamics, mechanisms of action and reported toxicity. Methods A search of the literature available on PubMed was done, using the keywords "ibogaine" and "noribogaine". The search criteria were "mechanism of action", "pharmacokinetics", "pharmacodynamics", "neurotransmitters", "toxicology", "toxicity", "cardiac", "neurotoxic", "human data", "animal data", "addiction", "anti-addictive", "withdrawal", "death" and "fatalities". The searches identified 382 unique references, of which 156 involved human data. Further research revealed 14 detailed toxicological case reports. Current Awareness in Clinical Toxicology is produced monthly for the American Academy of Clinical Toxicology by the Birmingham Unit of the UK National Poisons Information Service, with contributions from the Cardiff, Edinburgh, and Newcastle Units. The NPIS is commissioned by Public Health England Current Awareness in Clinical Toxicology Editors: Damian Ballam MSc and Allister Vale MD February 2016 Current Awareness in Clinical Toxicology is produced monthly for the American Academy of Clinical Toxicology by the Birmingham Unit of the UK National Poisons Information Service, with contributions from the Cardiff, Edinburgh, and Newcastle Units.
    [Show full text]
  • Biased Signaling by Endogenous Opioid Peptides
    Biased signaling by endogenous opioid peptides Ivone Gomesa, Salvador Sierrab,1, Lindsay Lueptowc,1, Achla Guptaa,1, Shawn Goutyd, Elyssa B. Margolise, Brian M. Coxd, and Lakshmi A. Devia,2 aDepartment of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; bDepartment of Physiology & Biophysics, Virginia Commonwealth University, Richmond, VA 23298; cSemel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095; dDepartment of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda MD 20814; and eDepartment of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94143 Edited by Susan G. Amara, National Institutes of Health, Bethesda, MD, and approved April 14, 2020 (received for review January 20, 2020) Opioids, such as morphine and fentanyl, are widely used for the possibility that endogenous opioid peptides could vary in this treatment of severe pain; however, prolonged treatment with manner as well (13). these drugs leads to the development of tolerance and can lead to For opioid receptors, studies showed that mice lacking opioid use disorder. The “Opioid Epidemic” has generated a drive β-arrestin2 exhibited enhanced and prolonged morphine-mediated for a deeper understanding of the fundamental signaling mecha- antinociception, and a reduction in side-effects, such as devel- nisms of opioid receptors. It is generally thought that the three opment of tolerance and acute constipation (15, 16). This led to types of opioid receptors (μ, δ, κ) are activated by endogenous studies examining whether μOR agonists exhibit biased signaling peptides derived from three different precursors: Proopiomelano- (17–20), and to the identification of agonists that preferentially cortin, proenkephalin, and prodynorphin.
    [Show full text]
  • BBC Program 2016.Pages
    March 5-6, 2016 La Quinta Inn & Suites Medical Center San Antonio, TX Behavior, Biology, and Chemistry: Translational Research in Addiction BBC 2016 BBC Publications BBC 2011 Stockton Jr SD and Devi LA (2012) Functional relevance of μ–δ opioid receptor heteromerization: A Role in novel signaling and implications for the treatment of addiction disorders: From a symposium on new concepts in mu-opioid pharmacology. Drug and Alcohol Dependence Mar 1;121(3):167-72. doi: 10.1016/j.drugalcdep.2011.10.025. Epub 2011 Nov 23 Traynor J (2012) μ-Opioid receptors and regulators of G protein signaling (RGS) proteins: From a sym- posium on new concepts in mu-opioid pharmacology. Drug and Alcohol Dependence Mar 1;121(3): 173-80. doi: 10.1016/j.drugalcdep.2011.10.027. Epub 2011 Nov 29 Lamb K, Tidgewell K, Simpson DS, Bohn LM and Prisinzano TE (2012) Antinociceptive effects of herkinorin, a MOP receptor agonist derived from salvinorin A in the formalin test in rats: New concepts in mu opioid receptor pharmacology: From a symposium on new concepts in mu-opioid pharma- cology. Drug and Alcohol Dependence Mar 1;121(3):181-8. doi: 10.1016/j.drugalcdep.2011.10.026. Epub 2011 Nov 26 Whistler JL (2012) Examining the role of mu opioid receptor endocytosis in the beneficial and side-ef- fects of prolonged opioid use: From a symposium on new concepts in mu-opioid pharmacology. Drug and Alcohol Dependence Mar 1;121(3):189-204. doi: 10.1016/j.drugalcdep.2011.10.031. Epub 2012 Jan 9 BBC 2012 Zorrilla EP, Heilig M, de Wit, H and Shaham Y (2013) Behavioral, biological, and chemical perspectives on targeting CRF1 receptor antagonists to treat alcoholism.
    [Show full text]
  • Differential Regulation of Serotonin 2A Receptor Responsiveness by Agonist
    Differential regulation of serotonin 2A receptor responsiveness by agonist- directed interactions with arrestin2 DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Cullen Laura Schmid, B.S. Neuroscience Graduate Studies Program The Ohio State University 2011 Dissertation Committee: Laura M. Bohn, Co-advisor Georgia A. Bishop, Co-advisor Candice C. Askwith Wolfgang Sadee Copyright by Cullen Laura Schmid 2011 Abstract The G protein-coupled, serotonin 2A (5-HT2A) receptor is a major drug target for the treatment of a number of mental health disorders, including schizophrenia, anxiety and depression. In addition to modulating several of the physiological effects of the neurotransmitter serotonin, activation of the 5-HT2A receptor mediates the psychotomimetic effects of serotonergic hallucinogenic drugs, such as lysergic acid diethylamide (LSD), 2,5-dimethoxy-4-iodoamphetamine (DOI) and 5-methoxy-N,N- dimethyltryptamine (5-MeO-DMT). Though hallucinogens are agonists at the 5-HT2A receptor, not all 5-HT2A receptor agonists induce hallucinations in humans, including the endogenous ligand serotonin. Therefore, the activation of the 5-HT2A receptor can result in different biological responses depending upon the chemical nature of the ligand, a concept that has been referred to as “functional selectivity.” One way in which ligands can induce differential signaling at GPCRs is through interactions with arrestins, which can act to dampen or facilitate receptor signaling cascades or mediate the internalization of receptors into intracellular vesicles. The overarching hypothesis of this dissertation is that the interaction between the regulatory protein, arrestin2, and the 5-HT2A receptor is a critical point in the divergence of agonist-directed 5-HT2A receptor responsiveness.
    [Show full text]
  • Fiona Bull Thesis Final Dec15.Pdf
    University of Dundee DOCTOR OF PHILOSOPHY Targeting Opioid Receptor Signal Transduction to Produce Sustained Analgesia Bull, Fiona A. Award date: 2015 Link to publication General rights Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal Take down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Download date: 04. Oct. 2021 Targeting Opioid Receptor Signal Transduction to Produce Sustained Analgesia Fiona A Bull BSc(Hons), MBChB, FRCA Thesis submitted in candidature for the degree of Doctor of Philosophy, University of Dundee, November 2015. i Table of Contents List of Tables -------------------------------------------------------------------------------------- vi List of Figures ------------------------------------------------------------------------------------ vii Abbreviations ----------------------------------------------------------------------------------- xii Acknowledgements
    [Show full text]
  • February, 2009
    NIDA - Director's Report - February, 2009 NIDA Home > Publications > Director's Reports Director's Report to the National Advisory Council on Drug Abuse - February, 2009 Index Research Findings Basic Neurosciences Research Basic Behavioral Research Behavioral and Brain Development Research Clinical Neuroscience Research Epidemiology and Etiology Research Prevention Research Research on Behavioral and Combined Treatments for Drug Abuse Research on Pharmacotherapies for Drug Abuse Research on Medical Consequences of Drug Abuse and Co-Occurring Infections (HIV/AIDS, HCV) Services Research Clinical Trials Network Research International Research Intramural Research Program Activities Extramural Policy and Review Activities Congressional Affairs International Activities Meetings and Conferences Media and Education Activities Planned Meetings Publications Staff Highlights Grantee Honors https://archives.drugabuse.gov/DirReports/DirRep209/Default.html[11/17/16, 11:14:00 PM] NIDA - Director's Report - February, 2009 Archive Home | Accessibility | Privacy | FOIA (NIH) | Current NIDA Home Page The National Institute on Drug Abuse (NIDA) is part of the National Institutes of Health (NIH) , a component of the U.S. Department of Health and Human Services. Questions? _ See our Contact Information. https://archives.drugabuse.gov/DirReports/DirRep209/Default.html[11/17/16, 11:14:00 PM] NIDA - Director's Report - February, 2009 NIDA Home > Publications > Director's Reports > February, 2009 Index Director's Report to the National Advisory Council on Drug Abuse - February, 2009 Index Research Findings - Basic Neuroscience Research Research Findings AMPA Glutamate Receptors Mediate Time Dependent Cue-Induced Cross-Divisional Research Craving for Cocaine Basic Neurosciences Research Increased time dependent craving for cocaine is frequently seen among cocaine addicts following abstinence and may account for relapse after periods of Basic Behavioral Research abstinence.
    [Show full text]
  • Copyright © Keith M. Olson 2017 a Dissertation Submitted to The
    Atypical Opioid Interactions – Development of Selective Mu-Delta Heterodimer Antagonists, Clinical Opioids at Non-Mu Pain Targets and Endogenous Biased Signaling Item Type text; Electronic Dissertation Authors Olson, Keith Mathew Publisher The University of Arizona. Rights Copyright © is held by the author. Digital access to this material is made possible by the University Libraries, University of Arizona. Further transmission, reproduction or presentation (such as public display or performance) of protected items is prohibited except with permission of the author. Download date 05/10/2021 19:11:41 Link to Item http://hdl.handle.net/10150/626669 ATYPICAL OPIOID INTERACTIONS – DEVELOPMENT OF SELECTIVE MU-DELTA HETERODIMER ANTAGONISTS, CLINICAL OPIOIDS AT NON-MU PAIN TARGETS AND ENDOGENOUS BIASED SIGNALING by Keith M. Olson ___________________ Copyright © Keith M. Olson 2017 A Dissertation Submitted to the Faculty of the DEPARTMENT OF CHEMISTRY AND BIOCHEMISTRY In Partial Fulfillment of the Requirements For the Degree of DOCTOR OF PHILOSOPHY WITH A MAJOR IN BIOCHEMISTRY In the Graduate College THE UNIVERSITY OF ARIZONA 2017 2 3 STATEMENT BY AUTHOR This dissertation has been submitted in partial fulfillment of requirements for an advanced degree at the University of Arizona and is deposited in the University Library to be made available to borrowers under rules of the Library. Brief quotations from this dissertation are allowable without special permission, provided that accurate acknowledgment of source is made. Requests for permission for extended quotation from or reproduction of this manuscript in whole or in part may be granted by the head of the major department or the Dean of the Graduate College when in his or her judgment the proposed use of the material is in the interests of scholarship.
    [Show full text]
  • (Herkinorin) and Internalizing (DAMGO) Μ-Opioid
    SYNAPSE 61:166–175 (2007) A Comparison of Noninternalizing (Herkinorin) and Internalizing (DAMGO) m-Opioid Agonists on Cellular Markers Related to Opioid Tolerance and Dependence HENG XU,1 JOHN S. PARTILLA,1 XIAOYING WANG,1 JOHN M. RUTHERFORD,1 KEVIN TIDGEWELL,2 2 3,4 1 THOMAS E. PRISINZANO, LAURA M. BOHN, AND RICHARD B. ROTHMAN * 1Clinical Psychopharmacology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, DHHS, Baltimore, Maryland 2Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, Iowa 3Department of Pharmacology, Ohio State University College of Medicine and Public Health, Columbus, Ohio 4Department of Psychiatry, Ohio State University College of Medicine and Public Health, Columbus, Ohio KEY WORDS DAMGO; herkinorin; tolerance; dependence; G proteins ABSTRACT Previous studies established that Tyr-D-Ala-Gly-N-Me-Phe-Gly-ol (DAMGO) and (2S,4aR,6aR,7R,9S,10aS,10bR)-9-(Benzoyloxy)-2-(3-furanyl)dodecahy- dro-6a,10b-dimethyl-4,10-dioxo-2H-naphtho-[2,1-c]pyran-7-carboxylic acid methyl ester (herkinorin) are fully efficacious m-agonists. Herkinorin (HERK), unlike DAMGO, does not recruit b-arrestin and promote m-receptor internalization, even in cells that over express b-arrestin. We hypothesized that chronic HERK and DAMGO treatment will differentially affect cellular markers of tolerance and dependence. CHO cells express- ing the cloned human m-receptor were treated for 20 h with 10 mM DAMGO, HERK, morphine, or medium. Both DAMGO and HERK acted as full agonists in the [35S]- GTP-g-SbindingassaywithEMAX values of 230% and EC50 values of 12.8 and 92.5 nM, respectively.
    [Show full text]
  • Src Kinase Inhibition Attenuates Morphine Tolerance Without Affecting Reinforcement Or Psychomotor Stimulation
    Src Kinase Inhibition Attenuates Morphine Tolerance without Affecting Reinforcement or Psychomotor Stimulation Fiona A. Bull, Ph.D., Daniel T. Baptista-Hon, Ph.D., Claire Sneddon, B.Sc., Lisa Wright, B.Sc., Wendy Walwyn, Ph.D., Tim G. Hales, Ph.D. ABSTRACT Background: Prolonged opioid administration leads to tolerance characterized by reduced analgesic potency. Pain manage- ment is additionally compromised by the hedonic effects of opioids, the cause of their misuse. The multifunctional protein β-arrestin2 regulates the hedonic effects of morphine and participates in tolerance. These actions might reflect µ opioid recep- Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/127/5/878/380385/20171100_0-00028.pdf by guest on 26 September 2021 tor up-regulation through reduced endocytosis. β-Arrestin2 also recruits kinases to µ receptors. We explored the role of Src kinase in morphine analgesic tolerance, locomotor stimulation, and reinforcement in C57BL/6 mice. Methods: Analgesic (tail withdrawal latency; percentage of maximum possible effect, n = 8 to 16), locomotor (distance traveled, n = 7 to 8), and reinforcing (conditioned place preference, n = 7 to 8) effects of morphine were compared in wild-type, µ+/–, µ–/–, and β-arrestin2–/– mice. The influence of c-Src inhibitors dasatinib (n = 8) and PP2 (n = 12) was examined. Results: Analgesia in morphine-treated wild-type mice exhibited tolerance, declining by day 10 to a median of 62% maxi- mum possible effect (interquartile range, 29 to 92%). Tolerance was absent from mice receiving dasatinib. Tolerance was enhanced in µ+/– mice (34% maximum possible effect; interquartile range, 5 to 52% on day 5); dasatinib attenuated tolerance (100% maximum possible effect; interquartile range, 68 to 100%), as did PP2 (91% maximum possible effect; interquartile range, 78 to 100%).
    [Show full text]
  • Agonist-Selective Regulation of the Mu Opioid Receptor by Βarrestins
    Agonist-selective regulation of the mu opioid receptor by βarrestins DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Chad Edward Groer Graduate Program in Integrated Biomedical Science Program The Ohio State University 2010 Dissertation Committee: Professor Laura M. Bohn. Co-Advisor Professor Wolfgang Sadée, Co-Advisor Professor John Oberdick Professor Lane Wallace Copyright by Chad Edward Groer 2010 ABSTRACT Morphine and other opiates mediate their effects through activation of the mu opioid receptor (MOR). Activation of the MOR results in recruitment of regulatory proteins, arrestins, that can regulate how this receptor signals. In vivo studies suggest that disruption of βarrestin-mediated MOR regulation may enhance opiate-induced antinociception and reduce tolerance and certain unwanted side effects. Therefore, by understanding the cellular mechanisms by which this receptor is regulated, the development of analgesics which preserve the beneficial effects of opiates while eliminating unwanted side effects may be possible. In this dissertation we test the hypothesis that MOR agonists can bias MOR-arrestin interactions, and that arrestin recruitment profiles, in turn, may determine cellular responses evoked by these agonists. In the first data portion of this dissertation, we characterize several novel MOR agonists that are unable to promote βarrestin recruitment. Herkinorin is a moderately selective agonist at the MOR, based on the structure of a natural product, Salvinorin A. We find that herkinorin promotes very little MOR phosphorylation, does not recruit βarrestins, and does not induce receptor internalization in transfected cells. Herkinorin is unable to induce βarrestin recruitment or MOR internalization under conditions that facilitate receptor ii phosphorylation and subsequent arrestin recruitment with other agonists.
    [Show full text]
  • Salvia Divinorum and Salvinorin A: an Update on Pharmacology And
    Oliver Grundmann Stephen M. Phipps Salvia divinorum and Salvinorin A: An Update on Immo Zadezensky Veronika Butterweck Pharmacology and Analytical Methodology Review Abstract derstand and elucidate the various medicinal properties of the plant itself and to provide the legislative authorities with enough Salvia divinorum (Lamiaceae) has been used for centuries by the information to cast judgement on S. divinorum. Mazatecan culture and has gained popularity as a recreational drug in recent years. Its potent hallucinogenic effects seen in Key words case reports has triggered research and led to the discovery of Salvia divinorum ´ Lamiaceae ´ depression ´ opioid antagonist ´ the first highly selective non-nitrogenous k opioid receptor ago- pharmacokinetics ´ pharmacology ´ toxicology ´ salvinorin A nist salvinorin A. This review critically evaluates the reported pharmacological and toxicological properties of S. divinorum Abbreviations and one of its major compounds salvinorin A, its pharmacokinet- CNS: central nervous system ic profile, and the analytical methods developed so far for its de- FST: forced swimming test tection and quantification. Recent research puts a strong empha- i.t.: intrathecal sis on salvinorin A, which has been shown to be a selective opioid KOR: kappa opioid receptor antagonist and is believed to have further beneficial properties, LSD: lysergic acid diethylamide rather than the leaf extract of S. divinorum. Currently animal norBNI: norbinaltorphimine studies show a rapid onset of action and short distribution and p.o.: per os 1039 elimination half-lives as well as a lack of evidence of short- or s.c.: subcutaneous long-term toxicity. Salvinorin A seems to be the most promising ssp.: subspecies approach to new treatment options for a variety of CNS illnesses.
    [Show full text]