Ion Binding Site As a Mechanism for Positive Allosteric Modulation of the Mu-Opioid Receptor

Total Page:16

File Type:pdf, Size:1020Kb

Ion Binding Site As a Mechanism for Positive Allosteric Modulation of the Mu-Opioid Receptor + Disruption of the Na ion binding site as a mechanism for positive allosteric modulation of the mu-opioid receptor Kathryn E. Livingston and John R. Traynor1 Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109 Edited by Leslie Lars Iversen, University of Oxford, Oxford, United Kingdom, and approved November 13, 2014 (received for review August 6, 2014) Positive allosteric modulation of the mu-opioid receptor (MOPr), of in vitro and in vivo literature describing allostery at GPCRs the site of action of all clinically used opioids, represents a poten- (9–11). In contrast, apart from our initial description of mu-PAMs, tial approach for the management of pain. We recently reported very little is known about allostery at MOPr. on positive allosteric modulators of MOPr (mu-PAMs), a class A G Allosteric modulators exhibit probe dependence, meaning they protein coupled receptor (GPCR). This study was designed to show disparate effects depending on the agonist bound to the examine the mechanism of allostery by comparing the degree to orthosteric site (12). A striking example of this is LY2033298, which opioid ligand structure governs modulation. To do this we a PAM of the muscarinic acetylcholine receptors M2 and M4. examined the interaction of the mu-PAM, BMS-986122, with LY2033298 increases the affinity of the agonist oxotremorine, a chemically diverse range of MOPr orthosteric ligands. Generally, while having no effect on the binding of the agonists pilocarpine for full agonists BMS-986122 enhanced the binding affinity and and McN-A-343 (13). Currently, it is not known if all opioid potency to activate G protein with no alteration in the maximal agonists are equally sensitive to the PAM effect of BMS-986122, effect. In contrast, lower efficacy agonists including morphine nor the mechanism underlying the allosteric modulation. Our were insensitive to alterations in binding affinity and showed little initial characterization showed that BMS-986122 causes a shift 2 4 to no change in potency to stimulate G protein. Instead, there was in the potency of the agonist DAMGO ([D-Ala , N-MePhe , an increase in maximal G protein stimulation. Antagonists were Gly-ol]-enkephalin), but increases the maximal stimulation of G unresponsive to the modulatory effects of BMS-986122. Sodium is protein by morphine (1). Opioid ligands are extremely diverse, a known endogenous allosteric modulator of MOPr and alters ranging from the 31-amino acid endogenous peptide β-endorphin to orthosteric agonist affinity and efficacy. The sensitivity of an small alkaloids like morphine. Therefore, this study sought to an- orthosteric ligand to BMS-986122 was strongly correlated with its swer two questions: (i) does BMS-986122 show probe dependence sensitivity to NaCl. In addition, BMS-986122 decreased the ability for the orthosteric ligand? And (ii) if probe dependence is seen, of NaCl to modulate agonist binding in an allosteric fashion. what is the mechanistic basis for this? Overall, BMS-986122 displayed marked probe dependence that To address these questions we examined the effect of BMS- was based upon the efficacy of the orthosteric ligand and can be 986122 on the MOPr properties of a wide range of opioid ligands PHARMACOLOGY explained using the Monod–Wyman–Changeux two-state model + from endogenous peptides to small molecules (Fig. S2). The of allostery. Furthermore, disruption of the Na ion binding site results reveal that the PAM effects of BMS-986122 are de- may represent a common mechanism for allosteric modulation of pendent on the efficacy of the orthosteric ligand and not on the class A GPCRs. structure per se. We find a strong correlation between the pos- + itive action of BMS-986122 and the negative action of Na ions endogenous opioids | opiates | GTPgammaS | ligand binding | receptor states Significance he mu-opioid receptor (MOPr) is the site of action of all Tclinically used opioid drugs. MOPr is a class A G protein- Morphine and related compounds are the gold standard for the coupled receptor (GPCR) that activates heterotrimeric Gi/o management of pain. Such drugs bind to the orthosteric site on proteins. Clinically used opioid agonists bind to the orthosteric the mu-opioid receptor (MOPr), a G protein-coupled receptor site on MOPr and although they are efficacious at causing pain (GPCR). We have proposed that targeting an allosteric site on relief, have a number of unwanted side effects resulting from MOPr could result in improved pain management and have direct MOPr activation. We have recently discovered and pre- reported positive allosteric modulators (PAMs) of MOPr. High- + sented a preliminary characterization of positive allosteric modu- resolution X-ray structures have identified a Na binding site + lators of MOPr (mu-PAMs) and are currently pursuing the idea on multiple GPCRs and have shown how bound Na stabilizes that mu-PAMs could be a viable way to manage pain (1, 2). The inactive receptor states. Here we demonstrate that PAM ac- ligand BMS-986122 (Fig. S1) represents the most active mu-PAM tivity at MOPr allosterically disrupts Na+ binding, thereby + currently identified. It was discovered in a high-throughput screen forming an active-state receptor. The Na binding site is highly for its ability to enhance the recruitment of β-arrestin to MOPr conserved across class A GPCRs so this may represent a funda- by the agonist endomorphin-1. Although having little agonist mental mechanism of allosteric modulation. activity on its own, this modulator has the ability to enhance the affinity, potency, and/or maximal response of MOPr agonists. In A preliminary report of some of the findings presented in this paper was given at the 45th annual meeting of the International Narcotics Research Conference, July 13–18, the same systems, BMS-986122 has no activity when the delta 2014, Montreal, Canada. opioid receptor (DOPr) is expressed, indicating the importance Author contributions: K.E.L. and J.R.T. designed research; K.E.L. performed research; of MOPr for BMS-986122 activity. K.E.L. and J.R.T. analyzed data; and K.E.L. and J.R.T. wrote the paper. The study of allosteric modulation of GPCRs has recently gained The authors declare no conflict of interest. momentum (3) and represents a relatively unexplored avenue for This article is a PNAS Direct Submission. drug development (4, 5). Allosteric modulators have been dis- 1To whom correspondence should be addressed. Email: [email protected]. covered for several GPCRs including the muscarinic, cannabinoid, This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. and metabotropic glutamate receptors (6–8) with a growing body 1073/pnas.1415013111/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1415013111 PNAS | December 23, 2014 | vol. 111 | no. 51 | 18369–18374 Downloaded by guest on September 25, 2021 to inhibit agonist binding. Moreover, we also show that BMS- Table 1. Binding affinity of MOPr ligands in the presence or + 986122 allosterically inhibits the ability of Na ions to reduce absence of BMS-986122 agonist binding. The PAM effect of BMS-986122 can conse- + Ligand Ki(Veh), nM Ki(BMS), nM Ki(Veh)/Ki(BMS) quently be explained by an inhibition of the ability of Na ions to stabilize the inactive state of the receptor, thereby allowing the Peptides receptor to shift to an active conformation. Thus, the mechanism β-Endorphin 194 ± 13 47 ± 8*** 4.1 ± ± of positive allosteric modulation can be simply explained by the Endomorphin-1 104 32 17 8* 6.1 ± ± two-state Monod–Wyman–Changeux model of allosterism (14). Leu-Enk 664 67 100 15** 6.6 Met-Enk 423 ± 133 63 ± 16* 6.7 Results Small molecules ± ± We first investigated the effects of a maximally effective con- Buprenorphine 0.5 0.1 0.4 0.2 1.3 ± ± centration (10 μM; ref. 1) of BMS-986122 on the MOPr activity Etorphine 2.4 0.3 1.5 0.8 1.6 ± ± of a range of endogenous opioid peptides (Fig. S2A). Using cell Fentanyl 222 34 89 25* 2.5 Loperamide 215 ± 54 14 ± 2* 15.4 membrane homogenates prepared from C6 glioma cells stably (RS)-Methadone 1076 ± 85 100 ± 4*** 10.7 expressing MOPr (C6MOPr) (15), we performed radioligand ± ± 3 (R)-Methadone 382 10 36 10*** 10.6 competition binding assays using H-diprenorphine (DPN, an ± ± γ (S)-Methadone 6,358 2,065 896 35* 7.1 opioid antagonist) in the presence of GTP S and NaCl to gen- Morphine 163 ± 18 143 ± 42 1.1 erate an inactive receptor state known to predominate in native Naloxone 2.5 ± 0.7 3.4 ± 1.0 0.7 membranes (16, 17). As shown in Fig. 1A and Table 1, BMS- 986122 caused an approximate sixfold enhancement in the Affinity (Ki values) were determined by competitive displacement of 3 affinity of both methionine-enkephalin (Met-Enk) and leucine- H-DPN binding from C6MOPr cell membranes in a buffer containing 10 μM γ enkephalin (Leu-Enk). A similar increase in affinity was seen for GTP S and 100 mM NaCl as described in Methods, in the absence or presence of 10 μM BMS-986122. *P < 0.05, **P < 0.01, ***P < 0.001 compared with control the smaller putative endogenous peptide endomorphin-1 (18). In (vehicle) data by Student t test. Data shown are means ± SEM of three to five independent experiments each in duplicate. addition to enhancing its affinity to bind MOPr, BMS-986122 caused a leftward shift in the concentration-response for Met- Enk to activate G protein, with no alteration in the maximal response (Emax), as measured by GTPγ35S binding in membrane homogenates (Fig. 1C and Table 2); an effect also seen with Leu-Enk and endomorphin-1 (Table 2). The endogenous opioid β-endorphin, a much larger 31-amino acid peptide, was also modulated by BMS-986122 with rightward shifts in both the af- finity (fourfold; Table 1) and potency (sixfold; Table 2) to acti- vate G protein.
Recommended publications
  • A,-, and P-Opioid Receptor Agonists on Excitatory Transmission in Lamina II Neurons of Adult Rat Spinal Cord
    The Journal of Neuroscience, August 1994, 74(E): 4965-4971 Inhibitory Actions of S,-, a,-, and p-Opioid Receptor Agonists on Excitatory Transmission in Lamina II Neurons of Adult Rat Spinal Cord Steven R. Glaum,’ Richard J. Miller,’ and Donna L. Hammond* Departments of lPharmacoloaical and Phvsioloqical Sciences and ‘Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois 60637 . This study examined the electrophysiological consequences tor in rat spinal cord and indicate that activation of either of selective activation of 6,-, 6,-, or r-opioid receptors using 6,- or Qopioid receptors inhibits excitatory, glutamatergic whole-cell recordings made from visually identified lamina afferent transmission in the spinal cord. This effect may me- II neurons in thin transverse slices of young adult rat lumbar diate the ability of 6, or 6, receptor agonists to produce an- spinal cord. Excitatory postsynaptic currents (EPSCs) or po- tinociception when administered intrathecally in the rat. tentials (EPSPs) were evoked electrically at the ipsilateral [Key words: DPDPE, deltorphin, spinal cord slice, EPSP, dorsal root entry zone after blocking inhibitory inputs with 6-opioid receptor, DAMGO, naltriben, 7-benzylidene- bicuculline and strychnine, and NMDA receptors with o-2- naltrexone (BNTX), naloxone] amino+phosphonopentanoic acid. Bath application of the p receptor agonist [D-Ala2, KMePhe4, Gly5-ollenkephalin (DAMGO) or the 6, receptor agonist [D-Pen2, o-PerF]en- The dorsal horn of the spinal cord is an important site for the kephalin (DPDPE) produced a log-linear, concentration-de- production of antinociception by K- and 6-opioid receptor ag- pendent reduction in the amplitude of the evoked EPSP/ onists (Yaksh, 1993).
    [Show full text]
  • 1 Title Page Pharmacological Comparison of Mitragynine and 7
    JPET Fast Forward. Published on December 31, 2020 as DOI: 10.1124/jpet.120.000189 This article has not been copyedited and formatted. The final version may differ from this version. JPET-AR-2020-000189R1: Obeng et al. Pharmacological Comparison of Mitragynine and 7-Hydroxymitragynine: In Vitro Affinity and Efficacy for Mu-Opioid Receptor and Morphine-Like Discriminative-Stimulus Effects in Rats. Title Page Pharmacological Comparison of Mitragynine and 7-Hydroxymitragynine: In Vitro Affinity and Efficacy for Mu-Opioid Receptor and Opioid-Like Behavioral Effects in Rats Samuel Obeng1,2,#, Jenny L. Wilkerson1,#, Francisco León2, Morgan E. Reeves1, Luis F. Restrepo1, Lea R. Gamez-Jimenez1, Avi Patel1, Anna E. Pennington1, Victoria A. Taylor1, Nicholas P. Ho1, Tobias Braun1, John D. Fortner2, Morgan L. Crowley2, Morgan R. Williamson1, Victoria L.C. Pallares1, Marco Mottinelli2, Carolina Lopera-Londoño2, Christopher R. McCurdy2, Lance R. McMahon1, and Takato Hiranita1* Downloaded from Departments of Pharmacodynamics1 and Medicinal Chemistry2, College of Pharmacy, University of Florida jpet.aspetjournals.org #These authors contributed equally Recommended section: Behavioral Pharmacology at ASPET Journals on September 29, 2021 Correspondence: *Takato Hiranita, Ph.D. Department of Pharmacodynamics, College of Pharmacy, University of Florida 1345 Center Drive, Gainesville, FL 32610 Email: [email protected] Phone: +1.352.294.5411 Fax: +1.352.273.7705 Keywords: kratom, mitragynine, 7-hydroxymitragynine, morphine, opioid, discriminative stimulus Conflicts of Interests or Disclaimers: None Manuscript statistics: Number of text pages: 56 Number of tables: 7 Number of figures: 10 Number of supplementary Tables: 4 Number of supplementary figures: 7 1 JPET Fast Forward. Published on December 31, 2020 as DOI: 10.1124/jpet.120.000189 This article has not been copyedited and formatted.
    [Show full text]
  • Heterodimerization of Μ and Δ Opioid Receptors: a Role in Opiate Synergy
    The Journal of Neuroscience, 2000, Vol. 20 RC110 1of5 Heterodimerization of ␮ and ␦ Opioid Receptors: A Role in Opiate Synergy I. Gomes, B. A. Jordan, A. Gupta, N. Trapaidze, V. Nagy, and L. A. Devi Departments of Pharmacology and Anesthesiology, New York University School of Medicine, New York, New York 10016 Opiate analgesics are widely used in the treatment of severe ␮-selective ligands results in a significant increase in the bind- pain. Because of their importance in therapy, different strate- ing of a ␦ receptor agonist. This robust increase is also seen in gies have been considered for making opiates more effective SKNSH cells that endogenously express both ␮ and ␦ recep- while curbing their liability to be abused. Although most opiates tors. Furthermore, we find that a ␦ receptor antagonist en- exert their analgesic effects primarily via ␮ opioid receptors, a hances both the potency and efficacy of the ␮ receptor signal- number of studies have shown that ␦ receptor-selective drugs ing; likewise a ␮ antagonist enhances the potency and efficacy can enhance their potency. The molecular basis for these find- of the ␦ receptor signaling. A combination of agonists (␮ and ␦ ings has not been elucidated previously. In the present study, receptor selective) also synergistically binds and potentiates we examined whether heterodimerization of ␮ and ␦ receptors signaling by activating the ␮–␦ heterodimer. Taken together, could account for the cross-modulation previously observed these studies show that heterodimers exhibit distinct ligand between these two receptors. We find that co-expression of ␮ binding and signaling characteristics. These findings have im- and ␦ receptors in heterologous cells followed by selective portant clinical ramifications and may provide new foundations immunoprecipitation results in the isolation of ␮–␦ het- for more effective therapies.
    [Show full text]
  • NIDA Drug Supply Program Catalog, 25Th Edition
    RESEARCH RESOURCES DRUG SUPPLY PROGRAM CATALOG 25TH EDITION MAY 2016 CHEMISTRY AND PHARMACEUTICS BRANCH DIVISION OF THERAPEUTICS AND MEDICAL CONSEQUENCES NATIONAL INSTITUTE ON DRUG ABUSE NATIONAL INSTITUTES OF HEALTH DEPARTMENT OF HEALTH AND HUMAN SERVICES 6001 EXECUTIVE BOULEVARD ROCKVILLE, MARYLAND 20852 160524 On the cover: CPK rendering of nalfurafine. TABLE OF CONTENTS A. Introduction ................................................................................................1 B. NIDA Drug Supply Program (DSP) Ordering Guidelines ..........................3 C. Drug Request Checklist .............................................................................8 D. Sample DEA Order Form 222 ....................................................................9 E. Supply & Analysis of Standard Solutions of Δ9-THC ..............................10 F. Alternate Sources for Peptides ...............................................................11 G. Instructions for Analytical Services .........................................................12 H. X-Ray Diffraction Analysis of Compounds .............................................13 I. Nicotine Research Cigarettes Drug Supply Program .............................16 J. Ordering Guidelines for Nicotine Research Cigarettes (NRCs)..............18 K. Ordering Guidelines for Marijuana and Marijuana Cigarettes ................21 L. Important Addresses, Telephone & Fax Numbers ..................................24 M. Available Drugs, Compounds, and Dosage Forms ..............................25
    [Show full text]
  • Self-Administration of Morphine, DAMGO, and DPDPE Into the Ventral Tegmental Area of Rats
    The Journal of Neuroscmce, April 1994. 74(4). 1978-1984 Self-Administration of Morphine, DAMGO, and DPDPE into the Ventral Tegmental Area of Rats- Darragh P. Devine and Roy A. Wise Center for Studies In Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada H3G lM8 Intracranial self-administration of K- and &opioid agonists ment associated with prior drug exposure. Conditioned place was demonstrated in male Long-Evans rats. Independent preferences are also observed after VTA microinjections of the groups were allowed to lever-press for ventral tegmental enkephalinasc inhibitor thiorphan (Glimcher et al., 1984). In area (VTA) microinfusions of morphine, the selective /* ag- addition, VT.4 microinjections ofmorphine lower the threshold onist [D-Ala*,N-Me-Phe4-GIy5-ol]-enkephalin (DAMGO), the for rewarding electrical stimulation of the lateral hypothalamus selective &agonist [o-Pen’,o-Pens]-enkephalin (DPDPE), or (Brockkamp et al., 1976; Jenck et al., 1987). ineffective drug vehicle. Morphine, DAMGO, and DPDPE were There arc at least three major types of opioid receptors (II, 6, each effective in establishing and maintaining lever-press- and K: Gilbert and Martin, 1976: Martin et al.. 1976; Lord et ing habits. Lever-pressing responses were extinguished al.: 1977; Paterson et al., 1983) that could potentially be in- during a session when vehicle was substituted for drug, and \-ol\,ed in the reuarding effects of opiates. The tritiated y agonist reinstated when drug reinforcement was reestablished. Thus, ‘H-DAMGO labels a dense population of FL-opioid receptors in it appears that VTA CL- and &opioid receptors are each in- the VTA (Quirion ct al.
    [Show full text]
  • Salvinorin a Analogues PR37 and PR38 Attenuate Compound 48
    British Journal of DOI:10.1111/bph.13212 www.brjpharmacol.org BJP Pharmacology RESEARCH PAPER Correspondence Jakub Fichna, Department of Biochemistry, Faculty of Medicine, Medical University of Salvinorin A analogues Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland. E-mail: jakub.fi[email protected] PR-37 and PR-38 attenuate ---------------------------------------------------------------- Received 18 January 2015 compound 48/80-induced Revised 26 May 2015 Accepted itch responses in mice 1 June 2015 M Salaga1, P R Polepally2, M Zielinska1, M Marynowski1, A Fabisiak1, N Murawska1, K Sobczak1, M Sacharczuk3,JCDoRego4, B L Roth5, J K Zjawiony2 and J Fichna1 1Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland, 2Department of BioMolecular Sciences, Division of Pharmacognosy and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, USA, 3Department of Molecular Cytogenetic, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzebiec, Poland, 4Platform of Behavioural Analysis (SCAC), Institute for Research and Innovation in Biomedicine (IRIB), Faculty of Medicine & Pharmacy, University of Rouen, Rouen Cedex, France, and 5Department of Pharmacology, Division of Chemical Biology and Medicinal Chemistry, Medical School, NIMH Psychoactive Drug Screening Program, University of North Carolina, Chapel Hill, NC, USA BACKGROUND AND PURPOSE The opioid system plays a crucial role in several physiological processes in the CNS and in the periphery. It has also been shown that selective opioid receptor agonists exert potent inhibitory action on pruritus and pain. In this study we examined whether two analogues of Salvinorin A, PR-37 and PR-38, exhibit antipruritic properties in mice. EXPERIMENTAL APPROACH To examine the antiscratch effect of PR-37 and PR-38 we used a mouse model of compound 48/80-induced pruritus.
    [Show full text]
  • Five Decades of Research on Opioid Peptides: Current Knowledge and Unanswered Questions
    Molecular Pharmacology Fast Forward. Published on June 2, 2020 as DOI: 10.1124/mol.120.119388 This article has not been copyedited and formatted. The final version may differ from this version. File name: Opioid peptides v45 Date: 5/28/20 Review for Mol Pharm Special Issue celebrating 50 years of INRC Five decades of research on opioid peptides: Current knowledge and unanswered questions Lloyd D. Fricker1, Elyssa B. Margolis2, Ivone Gomes3, Lakshmi A. Devi3 1Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; E-mail: [email protected] 2Department of Neurology, UCSF Weill Institute for Neurosciences, 675 Nelson Rising Lane, San Francisco, CA 94143, USA; E-mail: [email protected] 3Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, Annenberg Downloaded from Building, One Gustave L. Levy Place, New York, NY 10029, USA; E-mail: [email protected] Running Title: Opioid peptides molpharm.aspetjournals.org Contact info for corresponding author(s): Lloyd Fricker, Ph.D. Department of Molecular Pharmacology Albert Einstein College of Medicine 1300 Morris Park Ave Bronx, NY 10461 Office: 718-430-4225 FAX: 718-430-8922 at ASPET Journals on October 1, 2021 Email: [email protected] Footnotes: The writing of the manuscript was funded in part by NIH grants DA008863 and NS026880 (to LAD) and AA026609 (to EBM). List of nonstandard abbreviations: ACTH Adrenocorticotrophic hormone AgRP Agouti-related peptide (AgRP) α-MSH Alpha-melanocyte stimulating hormone CART Cocaine- and amphetamine-regulated transcript CLIP Corticotropin-like intermediate lobe peptide DAMGO D-Ala2, N-MePhe4, Gly-ol]-enkephalin DOR Delta opioid receptor DPDPE [D-Pen2,D- Pen5]-enkephalin KOR Kappa opioid receptor MOR Mu opioid receptor PDYN Prodynorphin PENK Proenkephalin PET Positron-emission tomography PNOC Pronociceptin POMC Proopiomelanocortin 1 Molecular Pharmacology Fast Forward.
    [Show full text]
  • In Vitro Effects of Ligand Bias on Primate Mu Opioid Receptor
    International Journal of Molecular Sciences Article In Vitro Effects of Ligand Bias on Primate Mu Opioid Receptor Downstream Signaling Xiao Zhang 1, Shaurita D. Hutchins 2 , Bruce E. Blough 3 and Eric J. Vallender 1,2,4,* 1 Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA; [email protected] 2 Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; [email protected] 3 Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, USA; [email protected] 4 Tulane National Primate Research Center, Covington, LA 70433, USA * Correspondence: [email protected] Received: 13 May 2020; Accepted: 2 June 2020; Published: 3 June 2020 Abstract: Interest has emerged in biased agonists at the mu opioid receptor (MOR) as a possible means for maintaining potent analgesis with reduced side effect profiles. While approaches measuring in vitro biased agonism are used in the development of these compounds, their therapeutic utility will ultimately be determined by in vivo functional effects. Nonhuman primates (NHPs) are the most translational model for evaluating the behavioral effects of candidate medications, but biased signaling of these drugs at NHP MOR receptors has been unstudied. The goal of the current work was to characterize MOR ligand bias in rhesus macaques, focusing on agonists that have previously been reported to show different patterns of biased agonism in rodents and humans. Downstream signaling pathways that responded to MOR activation were identified using a luciferase reporter array. Concentration-response curves for specific pathways (cAMP, NF-kB, MAPK/JNK) were generated using six agonists previously reported to differ in terms of signaling bias at rodent and human MORs.
    [Show full text]
  • Pharmacological Characterization of Dezocine, a Potent Analgesic Acting As a Κ Partial Agonist and Μ Partial Agonist
    www.nature.com/scientificreports OPEN Pharmacological Characterization of Dezocine, a Potent Analgesic Acting as a κ Partial Agonist and μ Received: 6 November 2017 Accepted: 31 August 2018 Partial Agonist Published: xx xx xxxx Yu-Hua Wang1, Jing-Rui Chai2, Xue-Jun Xu2, Ru-Feng Ye2, Gui-Ying Zan2, George Yun-Kun Liu3, Jian-Dong Long2, Yan Ma4, Xiang Huang2, Zhi-Chao Xiao2, Hu Dong2 & Yu-Jun Wang2 Dezocine is becoming dominated in China market for relieving moderate to severe pain. It is believed that Dezocine’s clinical efcacy and little chance to provoke adverse events during the therapeutic process are mainly attributed to its partial agonist activity at the μ opioid receptor. In the present work, we comprehensively studied the pharmacological characterization of Dezocine and identifed that the analgesic efect of Dezocine was a result of action at both the κ and μ opioid receptors. We frstly found that Dezocine displayed preferential binding to μ opioid receptor over κ and δ opioid receptors. Dezocine, on its own, weakly stimulated G protein activation in cells expressing κ and μ receptors, but in the presence of full κ agonist U50,488 H and μ agonist DAMGO, Dezocine inhibited U50,488H- and DAMGO-mediated G protein activation, indicating that Dezocine was a κ partial agonist and μ partial agonist. Then the in intro results were verifed by in vivo studies in mice. We observed that Dezocine- produced antinociception was signifcantly inhibited by κ antagonist nor-BNI and μ antagonist β-FNA pretreatment, indicating that Dezocine-mediated antinociception was via both the κ and μ opioid receptors.
    [Show full text]
  • Nalbuphine Suppresses Breast Cancer Stem-Like Properties and Epithelial
    Yu et al. Journal of Experimental & Clinical Cancer Research (2019) 38:197 https://doi.org/10.1186/s13046-019-1184-1 RESEARCH Open Access Nalbuphine suppresses breast cancer stem- like properties and epithelial-mesenchymal transition via the AKT-NFκB signaling pathway Jiachuan Yu1,2†, Yuanyuan Luo2† and Qingping Wen1* Abstract Background: Cancer pain is a debilitating disorder of human breast cancer and a primary determinant of the poor quality of life, and relieving pain is fundamental strategy in the cancer treatment. However, opioid analgesics, like morphine and fentanyl, which are widely used in cancer pain treatment have been reported to enhance stem-like traits and epithelial-mesenchymal transition (EMT) of breast cancer cells. As such, it is vital to make the best choice of analgesic for breast cancer management. Methods: MTT assays and colony formation assays were performed to examine tumor cell proliferation upon nalbuphine treatment. RT-PCR, western blot, flow cytometry, sphere formation, immunohistochemistry, transwell assays, wound healing assays and mouse xenograft were used to assess the biological effects of nalbuphine treatment. Results: Nalbuphine inhibited breast cancer cell growth and tumorigenesis, with little effect on noncancerous breast cell lines. Nalbuphine suppressed cancer stem-like traits and EMT in both breast cancer cells and mouse xenograft tumor tissues. Additionally, activation of AKT reversed the nalbuphine-induced inhibition of cancer stem- like properties, tumorigenesis and EMT. Conclusions: Our results demonstrate a new role of nalbuphine in inhibiting cancer stem-like properties and EMT in addition to relieving pain, which suggests that nalbuphine may be effective in breast cancer treatment. Keywords: Nalbuphine, Breast cancer, Stem-like traits, Epithelial-mesenchymal transition, AKT-NFκB pathway Background induce cancer metastasis, angiogenesis, and drug resist- A large number of breast cancer patients suffer from ance.
    [Show full text]
  • Heterologous Desensitization of Opioid Receptors by Chemokines Inhibits Chemotaxis and Enhances the Perception of Pain
    Heterologous desensitization of opioid receptors by chemokines inhibits chemotaxis and enhances the perception of pain Imre Szabo*†, Xiao-Hong Chen†‡, Li Xin†‡, Martin W. Adler†‡, O. M. Z. Howard§, Joost J. Oppenheim§, and Thomas J. Rogers*†¶ Departments of *Microbiology and Immunology, ‡Pharmacology, and the †Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140; and §Laboratory of Molecular Immunoregulation, Division of Basic Sciences, National Cancer Institute–Frederick Cancer Research and Development Center, Frederick, MD 21702-1201 Communicated by Henry Metzger, National Institutes of Health, Bethesda, MD, May 23, 2002 (received for review May 1, 2002) The chemokines use G protein-coupled receptors to regulate the opioid pretreatment is consistent with the desensitization of migratory and proadhesive responses of leukocytes. Based on either CCR1 or CCR5, or both. This receptor crosstalk resulting observations that G protein-coupled receptors undergo heterolo- in heterologous desensitization and phosphorylation of some of gous desensitization, we have examined the ability of chemokines the chemokine receptors may contribute to the immunosuppres- to also influence the perception of pain by cross-desensitizing sive effects of the opioids. opioid G protein-coupled receptors function in vitro and in vivo.We Conversely, we have considered the possibility that one or find that the chemotactic activities of both ␮- and ␦-opioid recep- more of the chemokine receptors may desensitize the opioid tors are desensitized following activation of the chemokine recep- receptors. We hypothesized that prior exposure to chemokines tors CCR5, CCR2, CCR7, and CXCR4 but not of the CXCR1 or CXCR2 might result in heterologous desensitization of opioid receptors, receptors.
    [Show full text]
  • The Atypical Chemokine Receptor ACKR3/CXCR7 Is a Broad-Spectrum Scavenger for Opioid Peptides
    University of Southern Denmark The atypical chemokine receptor ACKR3/CXCR7 is a broad-spectrum scavenger for opioid peptides Meyrath, Max; Szpakowska, Martyna; Zeiner, Julian; Massotte, Laurent; Merz, Myriam P.; Benkel, Tobias; Simon, Katharina; Ohnmacht, Jochen; Turner, Jonathan D.; Krüger, Rejko; Seutin, Vincent; Ollert, Markus; Kostenis, Evi; Chevigné, Andy Published in: Nature Communications DOI: 10.1038/s41467-020-16664-0 Publication date: 2020 Document version: Final published version Document license: CC BY Citation for pulished version (APA): Meyrath, M., Szpakowska, M., Zeiner, J., Massotte, L., Merz, M. P., Benkel, T., Simon, K., Ohnmacht, J., Turner, J. D., Krüger, R., Seutin, V., Ollert, M., Kostenis, E., & Chevigné, A. (2020). The atypical chemokine receptor ACKR3/CXCR7 is a broad-spectrum scavenger for opioid peptides. Nature Communications, 11, [3033]. https://doi.org/10.1038/s41467-020-16664-0 Go to publication entry in University of Southern Denmark's Research Portal Terms of use This work is brought to you by the University of Southern Denmark. Unless otherwise specified it has been shared according to the terms for self-archiving. If no other license is stated, these terms apply: • You may download this work for personal use only. • 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 this open access version If you believe that this document breaches copyright please contact us providing details and we will investigate your claim. Please direct all enquiries to [email protected] Download date: 05. Oct. 2021 ARTICLE https://doi.org/10.1038/s41467-020-16664-0 OPEN The atypical chemokine receptor ACKR3/CXCR7 is a broad-spectrum scavenger for opioid peptides Max Meyrath1,9, Martyna Szpakowska 1,9, Julian Zeiner2, Laurent Massotte3, Myriam P.
    [Show full text]