1
|
El Daibani A, Madasu MK, Al-Hasani R, Che T. Limitations and potential of κOR biased agonists for pain and itch management. Neuropharmacology 2024; 258:110061. [PMID: 38960136 PMCID: PMC11968146 DOI: 10.1016/j.neuropharm.2024.110061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/20/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024]
Abstract
The concept of ligand bias is based on the premise that different agonists can elicit distinct responses by selectively activating the same receptor. These responses often determine whether an agonist has therapeutic or undesirable effects. Therefore, it would be highly advantageous to have agonists that specifically trigger the therapeutic response. The last two decades have seen a growing trend towards the consideration of ligand bias in the development of ligands to target the κ-opioid receptor (κOR). Most of these ligands selectively favor G-protein signaling over β-arrestin signaling to potentially provide effective pain and itch relief without adverse side effects associated with κOR activation. Importantly, the specific role of β-arrestin 2 in mediating κOR agonist-induced side effects remains unknown, and similarly the therapeutic and side-effect profiles of G-protein-biased κOR agonists have not been established. Furthermore, some drugs previously labeled as G-protein-biased may not exhibit true bias but may instead be either low-intrinsic-efficacy or partial agonists. In this review, we discuss the established methods to test ligand bias, their limitations in measuring bias factors for κOR agonists, as well as recommend the consideration of other systematic factors to correlate the degree of bias signaling and pharmacological effects. This article is part of the Special Issue on "Ligand Bias".
Collapse
Affiliation(s)
- Amal El Daibani
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Manish K Madasu
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ream Al-Hasani
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA.
| | - Tao Che
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
2
|
Hampsey E, Jelen L, Young AH. Aticaprant: (a κ-opioid receptor antagonist) for major depressive disorder. Expert Opin Emerg Drugs 2024; 29:193-204. [PMID: 38682267 DOI: 10.1080/14728214.2024.2345645] [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: 02/20/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION Major depression is a common, disabling mental health condition associated with the highest disease burden for any neuropsychiatric disorder worldwide, according to the WHO. Due to the imperfect efficacy and tolerability profiles of existing treatments, investigational compounds in novel treatment classes are needed. Opioid-receptor antagonists are a potential new class of treatments currently under investigation. AREAS COVERED Major depressive disorder is first overviewed. Existing treatments, both their mechanisms of action and their place within the antidepressant space, are discussed herein. Then, the profile of Aticaprant and the wider context of kappa-opioid antagonism for depression are discussed in focus. EXPERT OPINION Early evidence indicates that Aticaprant may possess desirable pharmacodynamic and pharmacokinetic properties. A lack of convincing efficacy data at the time of writing precludes any definitive statement on its potential as an antidepressant.
Collapse
Affiliation(s)
- Elliot Hampsey
- Centre for Affective Disorders, King's College London, London, UK
| | - Luke Jelen
- Centre for Affective Disorders, King's College London, London, UK
| | - Allan H Young
- Centre for Affective Disorders, King's College London, London, UK
- South London & Maudsley NHS Foundation Trust, London, UK
| |
Collapse
|
3
|
Guo X, Akanda N, Fiorino G, Nimbalkar S, Long CJ, Colón A, Patel A, Tighe PJ, Hickman JJ. Human IPSC-Derived PreBötC-Like Neurons and Development of an Opiate Overdose and Recovery Model. Adv Biol (Weinh) 2024; 8:e2300276. [PMID: 37675827 PMCID: PMC10921423 DOI: 10.1002/adbi.202300276] [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: 08/04/2023] [Indexed: 09/08/2023]
Abstract
Opioid overdose is the leading cause of drug overdose lethality, posing an urgent need for investigation. The key brain region for inspiratory rhythm regulation and opioid-induced respiratory depression (OIRD) is the preBötzinger Complex (preBötC) and current knowledge has mainly been obtained from animal systems. This study aims to establish a protocol to generate human preBötC neurons from induced pluripotent cells (iPSCs) and develop an opioid overdose and recovery model utilizing these iPSC-preBötC neurons. A de novo protocol to differentiate preBötC-like neurons from human iPSCs is established. These neurons express essential preBötC markers analyzed by immunocytochemistry and demonstrate expected electrophysiological responses to preBötC modulators analyzed by patch clamp electrophysiology. The correlation of the specific biomarkers and function analysis strongly suggests a preBötC-like phenotype. Moreover, the dose-dependent inhibition of these neurons' activity is demonstrated for four different opioids with identified IC50's comparable to the literature. Inhibition is rescued by naloxone in a concentration-dependent manner. This iPSC-preBötC mimic is crucial for investigating OIRD and combating the overdose crisis and a first step for the integration of a functional overdose model into microphysiological systems.
Collapse
Affiliation(s)
- Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Nesar Akanda
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Gabriella Fiorino
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Siddharth Nimbalkar
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Christopher J Long
- Hesperos Inc, 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Alisha Colón
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Aakash Patel
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Patrick J Tighe
- College of Medicine, Department of Anesthesiology, University of Florida, 1600 SW Archer Road, Gainesville, FL, 32610, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
- Hesperos Inc, 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| |
Collapse
|
4
|
The Opioid System in Depression. Neurosci Biobehav Rev 2022; 140:104800. [PMID: 35914624 PMCID: PMC10166717 DOI: 10.1016/j.neubiorev.2022.104800] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 12/16/2022]
Abstract
Opioid receptors are widely distributed throughout the brain and play an essential role in modulating aspects of human mood, reward, and well-being. Accumulating evidence indicates the endogenous opioid system is dysregulated in depression and that pharmacological modulators of mu, delta, and kappa opioid receptors hold potential for the treatment of depression. Here we review animal and clinical data, highlighting evidence to support: dysregulation of the opioid system in depression, evidence for opioidergic modulation of behavioural processes and brain regions associated with depression, and evidence for opioidergic modulation in antidepressant responses. We evaluate clinical trials that have examined the safety and efficacy of opioidergic agents in depression and consider how the opioid system may be involved in the effects of other treatments, including ketamine, that are currently understood to exert antidepressant effects through non-opioidergic actions. Finally, we explore key neurochemical and molecular mechanisms underlying the potential therapeutic effects of opioid system engagement, that together provides a rationale for further investigation into this relevant target in the treatment of depression.
Collapse
|
5
|
Quantitative Systems Pharmacology and Biased Agonism at Opioid Receptors: A Potential Avenue for Improved Analgesics. Int J Mol Sci 2022; 23:ijms23095114. [PMID: 35563502 PMCID: PMC9104178 DOI: 10.3390/ijms23095114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/01/2022] [Accepted: 05/02/2022] [Indexed: 11/25/2022] Open
Abstract
Chronic pain is debilitating and represents a significant burden in terms of personal and socio-economic costs. Although opioid analgesics are widely used in chronic pain treatment, many patients report inadequate pain relief or relevant adverse effects, highlighting the need to develop analgesics with improved efficacy/safety. Multiple evidence suggests that G protein-dependent signaling triggers opioid-induced antinociception, whereas arrestin-mediated pathways are credited with modulating different opioid adverse effects, thus spurring extensive research for G protein-biased opioid agonists as analgesic candidates with improved pharmacology. Despite the increasing expectations of functional selectivity, translating G protein-biased opioid agonists into improved therapeutics is far from being fully achieved, due to the complex, multidimensional pharmacology of opioid receptors. The multifaceted network of signaling events and molecular processes underlying therapeutic and adverse effects induced by opioids is more complex than the mere dichotomy between G protein and arrestin and requires more comprehensive, integrated, network-centric approaches to be fully dissected. Quantitative Systems Pharmacology (QSP) models employing multidimensional assays associated with computational tools able to analyze large datasets may provide an intriguing approach to go beyond the greater complexity of opioid receptor pharmacology and the current limitations entailing the development of biased opioid agonists as improved analgesics.
Collapse
|
6
|
Castro NCF, Silva IS, Cartágenes SC, Fernandes LMP, Ribera PC, Barros MA, Prediger RD, Fontes-Júnior EA, Maia CSF. Morphine Perinatal Exposure Induces Long-Lasting Negative Emotional States in Adult Offspring Rodents. Pharmaceutics 2021; 14:pharmaceutics14010029. [PMID: 35056925 PMCID: PMC8778186 DOI: 10.3390/pharmaceutics14010029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/28/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022] Open
Abstract
Psychoactive substances during pregnancy and lactation is a key problem in contemporary society, causing social, economic, and health disturbance. In 2010, about 30 million people used opioid analgesics for non-therapeutic purposes, and the prevalence of opioids use during pregnancy ranged from 1% to 21%, representing a public health problem. This study aimed to evaluate the long-lasting neurobehavioral and nociceptive consequences in adult offspring rats and mice exposed to morphine during intrauterine/lactation periods. Pregnant rats and mice were exposed subcutaneously to morphine (10 mg/kg/day) during 42 consecutive days (from the first day of pregnancy until the last day of lactation). Offspring were weighed on post-natal days (PND) 1, 5, 10, 15, 20, 30, and 60, and behavioral tasks (experiment 1) or nociceptive responses (experiment 2) were assessed at 75 days of age (adult life). Morphine-exposed female rats displayed increased spontaneous locomotor activity. More importantly, both males and female rats perinatally exposed to morphine displayed anxiety- and depressive-like behaviors. Morphine-exposed mice presented alterations in the nociceptive responses on the writhing test. This study showed that sex difference plays a role in pain threshold and that deleterious effects of morphine during pre/perinatal periods are nonrepairable in adulthood, which highlights the long-lasting clinical consequences related to anxiety, depression, and nociceptive disorders in adulthood followed by intrauterine and lactation morphine exposure.
Collapse
Affiliation(s)
- Nair C. F. Castro
- Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Universidade Federal do Pará, Belém 66075-900, Brazil; (N.C.F.C.); (I.S.S.); (S.C.C.); (P.C.R.); (M.A.B.); (E.A.F.-J.)
| | - Izabelle S. Silva
- Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Universidade Federal do Pará, Belém 66075-900, Brazil; (N.C.F.C.); (I.S.S.); (S.C.C.); (P.C.R.); (M.A.B.); (E.A.F.-J.)
| | - Sabrina C. Cartágenes
- Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Universidade Federal do Pará, Belém 66075-900, Brazil; (N.C.F.C.); (I.S.S.); (S.C.C.); (P.C.R.); (M.A.B.); (E.A.F.-J.)
| | - Luanna M. P. Fernandes
- Departamento de Ciências Morfológicas e Fisiológicas, Centro das Ciências Biológicas e da Saúde (CCBS), Universidade Estadual do Pará, Belém 66087-662, Brazil;
| | - Paula C. Ribera
- Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Universidade Federal do Pará, Belém 66075-900, Brazil; (N.C.F.C.); (I.S.S.); (S.C.C.); (P.C.R.); (M.A.B.); (E.A.F.-J.)
| | - Mayara A. Barros
- Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Universidade Federal do Pará, Belém 66075-900, Brazil; (N.C.F.C.); (I.S.S.); (S.C.C.); (P.C.R.); (M.A.B.); (E.A.F.-J.)
| | - Rui D. Prediger
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis 88040-900, Brazil;
| | - Enéas A. Fontes-Júnior
- Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Universidade Federal do Pará, Belém 66075-900, Brazil; (N.C.F.C.); (I.S.S.); (S.C.C.); (P.C.R.); (M.A.B.); (E.A.F.-J.)
| | - Cristiane S. F. Maia
- Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Universidade Federal do Pará, Belém 66075-900, Brazil; (N.C.F.C.); (I.S.S.); (S.C.C.); (P.C.R.); (M.A.B.); (E.A.F.-J.)
- Correspondence:
| |
Collapse
|
7
|
Stefanucci A, Iobbi V, Della Valle A, Scioli G, Pieretti S, Minosi P, Mirzaie S, Novellino E, Mollica A. In Silico Identification of Tripeptides as Lead Compounds for the Design of KOR Ligands. Molecules 2021; 26:4767. [PMID: 34443366 PMCID: PMC8399634 DOI: 10.3390/molecules26164767] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/21/2022] Open
Abstract
The kappa opioid receptor (KOR) represents an attractive target for the development of drugs as potential antidepressants, anxiolytics and analgesics. A robust computational approach may guarantee a reduction in costs in the initial stages of drug discovery, novelty and accurate results. In this work, a virtual screening workflow of a library consisting of ~6 million molecules was set up, with the aim to find potential lead compounds that could manifest activity on the KOR. This in silico study provides a significant contribution in the identification of compounds capable of interacting with a specific molecular target. The main computational techniques adopted in this experimental work include: (i) virtual screening; (ii) drug design and leads optimization; (iii) molecular dynamics. The best hits are tripeptides prepared via solution phase peptide synthesis. These were tested in vivo, revealing a good antinociceptive effect after subcutaneous administration. However, further work is due to delineate their full pharmacological profile, in order to verify the features predicted by the in silico outcomes.
Collapse
Affiliation(s)
- Azzurra Stefanucci
- Department of Pharmacy, University G. d’Annunzio Chieti, Via dei Vestini 31, 66100 Chieti, Italy; (A.S.); (A.D.V.); (G.S.)
| | - Valeria Iobbi
- Department of Pharmacy (DIFAR), University of Genova, 16128 Genova, Italy;
| | - Alice Della Valle
- Department of Pharmacy, University G. d’Annunzio Chieti, Via dei Vestini 31, 66100 Chieti, Italy; (A.S.); (A.D.V.); (G.S.)
| | - Giuseppe Scioli
- Department of Pharmacy, University G. d’Annunzio Chieti, Via dei Vestini 31, 66100 Chieti, Italy; (A.S.); (A.D.V.); (G.S.)
| | - Stefano Pieretti
- Centro Nazionale Ricerca e Valutazione Preclinica e Clinica dei Farmaci, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (S.P.); (P.M.)
| | - Paola Minosi
- Centro Nazionale Ricerca e Valutazione Preclinica e Clinica dei Farmaci, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (S.P.); (P.M.)
| | - Sako Mirzaie
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, 27 King’s College Circle, Toronto, ON M5S 1A1, Canada;
| | - Ettore Novellino
- NGN Healthcare, Via Nazionale Torrette, 207, 83013 Mercogliano, Italy;
| | - Adriano Mollica
- Department of Pharmacy, University G. d’Annunzio Chieti, Via dei Vestini 31, 66100 Chieti, Italy; (A.S.); (A.D.V.); (G.S.)
| |
Collapse
|
8
|
Abstract
Preclinical evidence has highlighted the importance of the μ-opioid peptide (MOP) receptor on primary afferents for both the analgesic actions of MOP receptor agonists, as well as the development of tolerance, if not opioid-induced hyperalgesia. There is also growing interest in targeting other opioid peptide receptor subtypes (δ-opioid peptide [DOP], κ-opioid peptide [KOP], and nociceptin/orphanin-FQ opioid peptide [NOP]) on primary afferents, as alternatives to MOP receptors, which may not be associated with as many deleterious side effects. Nevertheless, results from several recent studies of human sensory neurons indicate that although there are many similarities between rodent and human sensory neurons, there may also be important differences. Thus, the purpose of this study was to assess the distribution of opioid receptor subtypes among human sensory neurons. A combination of pharmacology, patch-clamp electrophysiology, Ca imaging, and single-cell semiquantitative polymerase chain reaction was used. Our results suggest that functional MOP-like receptors are present in approximately 50% of human dorsal root ganglion neurons. δ-opioid peptide-like receptors were detected in a subpopulation largely overlapping that with MOP-like receptors. Furthermore, KOP-like and NOP-like receptors are detected in a large proportion (44% and 40%, respectively) of human dorsal root ganglion neurons with KOP receptors also overlapping with MOP receptors at a high rate (83%). Our data confirm that all 4 opioid receptor subtypes are present and functional in human sensory neurons, where the overlap of DOP, KOP, and NOP receptors with MOP receptors suggests that activation of these other opioid receptor subtypes may also have analgesic efficacy.
Collapse
|
9
|
Paton KF, Biggerstaff A, Kaska S, Crowley RS, La Flamme AC, Prisinzano TE, Kivell BM. Evaluation of Biased and Balanced Salvinorin A Analogs in Preclinical Models of Pain. Front Neurosci 2020; 14:765. [PMID: 32792903 PMCID: PMC7385413 DOI: 10.3389/fnins.2020.00765] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/29/2020] [Indexed: 01/09/2023] Open
Abstract
In the search for safer, non-addictive analgesics, kappa opioid receptor (KOPr) agonists are a potential target, as unlike mu-opioid analgesics, they do not have abuse potential. Salvinorin A (SalA) is a potent and selective KOPr agonist, however, clinical utility is limited by the short duration of action and aversive side effects. Biasing KOPr signaling toward G-protein activation has been highlighted as a key cellular mechanism to reduce the side effects of KOPr agonists. The present study investigated KOPr signaling bias and the acute antinociceptive effects and side effects of two novel analogs of SalA, 16-Bromo SalA and 16-Ethynyl SalA. 16-Bromo SalA showed G-protein signaling bias, whereas 16-Ethynyl SalA displayed balanced signaling properties. In the dose-response tail-withdrawal assay, SalA, 16-Ethynyl SalA and 16-Bromo SalA were more potent than the traditional KOPr agonist U50,488, and 16-Ethynyl SalA was more efficacious. 16-Ethynyl SalA and 16-Bromo SalA both had a longer duration of action in the warm water tail-withdrawal assay, and 16-Ethynyl had greater antinociceptive effect in the hot-plate assay, compared to SalA. In the intraplantar 2% formaldehyde test, 16-Ethynyl SalA and 16-Bromo SalA significantly reduced both nociceptive and inflammatory pain-related behaviors. Moreover, 16-Ethynyl SalA and 16-Bromo SalA had no anxiogenic effects in the marble burying task, and 16-Bromo SalA did not alter behavior in the elevated zero maze. Overall, 16-Ethynyl SalA significantly attenuated acute pain-related behaviors in multiple preclinical models, while the biased KOPr agonist, 16-Bromo SalA, displayed modest antinociceptive effects, and lacked anxiogenic effects.
Collapse
Affiliation(s)
- Kelly F Paton
- School of Biological Sciences, Centre for Biodiscovery, Faculty of Science, Victoria University of Wellington, Wellington, New Zealand
| | - Andrew Biggerstaff
- School of Biological Sciences, Centre for Biodiscovery, Faculty of Science, Victoria University of Wellington, Wellington, New Zealand
| | - Sophia Kaska
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Rachel S Crowley
- Department of Medicinal Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS, United States
| | - Anne C La Flamme
- School of Biological Sciences, Centre for Biodiscovery, Faculty of Science, Victoria University of Wellington, Wellington, New Zealand.,Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Thomas E Prisinzano
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States.,Department of Medicinal Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS, United States
| | - Bronwyn M Kivell
- School of Biological Sciences, Centre for Biodiscovery, Faculty of Science, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
10
|
In Vitro Effects of Ligand Bias on Primate Mu Opioid Receptor Downstream Signaling. Int J Mol Sci 2020; 21:ijms21113999. [PMID: 32503269 PMCID: PMC7312292 DOI: 10.3390/ijms21113999] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/30/2020] [Accepted: 06/02/2020] [Indexed: 12/20/2022] Open
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-ĸB, MAPK/JNK) were generated using six agonists previously reported to differ in terms of signaling bias at rodent and human MORs. Using DAMGO as a reference ligand, relative cAMP, NF-ĸB and MAPK/JNK signaling by morphine, endomorphin-1, and TRV130 were found to be comparable between species. Further, the bias patterns of across ligands for NF-ĸB and MAPK/JNK were largely similar between species. There was a high degree of concordance between rhesus macaque and human MOR receptor signaling bias for all agonists tested, further demonstrating their utility for future translational behavioral studies.
Collapse
|
11
|
Bedini A, Di Cesare Mannelli L, Micheli L, Baiula M, Vaca G, De Marco R, Gentilucci L, Ghelardini C, Spampinato S. Functional Selectivity and Antinociceptive Effects of a Novel KOPr Agonist. Front Pharmacol 2020; 11:188. [PMID: 32210803 PMCID: PMC7066533 DOI: 10.3389/fphar.2020.00188] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/10/2020] [Indexed: 01/21/2023] Open
Abstract
Kappa opioid receptor (KOPr) agonists represent alternative analgesics for their low abuse potential, although relevant adverse effects have limited their clinical use. Functionally selective KOPr agonists may activate, in a pathway-specific manner, G protein-mediated signaling, that produces antinociception, over β-arrestin 2-dependent induction of p38MAPK, which preferentially contributes to adverse effects. Thus, functionally selective KOPr agonists biased toward G protein-coupled intracellular signaling over β-arrestin-2-mediated pathways may be considered candidate therapeutics possibly devoid of many of the typical adverse effects elicited by classic KOPr agonists. Nonetheless, the potential utility of functionally selective agonists at opioid receptors is still highly debated; therefore, further studies are necessary to fully understand whether it will be possible to develop more effective and safer analgesics by exploiting functional selectivity at KOPr. In the present study we investigated in vitro functional selectivity and in vivo antinociceptive effects of LOR17, a novel KOPr selective peptidic agonist that we synthesized. LOR17-mediated effects on adenylyl cyclase inhibition, ERK1/2, p38MAPK phosphorylation, and astrocyte cell proliferation were studied in HEK-293 cells expressing hKOPr, U87-MG glioblastoma cells, and primary human astrocytes; biased agonism was investigated via cAMP ELISA and β-arrestin 2 recruitment assays. Antinociception and antihypersensitivity were assessed in mice via warm-water tail-withdrawal test, intraperitoneal acid-induced writhing, and a model of oxaliplatin-induced neuropathic cold hypersensitivity. Effects of LOR17 on locomotor activity, exploratory activity, and forced-swim behavior were also assayed. We found that LOR17 is a selective, G protein biased KOPr agonist that inhibits adenylyl cyclase and activates early-phase ERK1/2 phosphorylation. Conversely to classic KOPr agonists as U50,488, LOR17 neither induces p38MAPK phosphorylation nor increases KOPr-dependent, p38MAPK-mediated cell proliferation in astrocytes. Moreover, LOR17 counteracts, in a concentration-dependent manner, U50,488-induced p38MAPK phosphorylation and astrocyte cell proliferation. Both U50,488 and LOR17 display potent antinociception in models of acute nociception, whereas LOR17 counteracts oxaliplatin-induced thermal hypersensitivity better than U50,488, and it is effective after single or repeated s.c. administration. LOR17 administered at a dose that fully alleviated oxaliplatin-induced thermal hypersensitivity did not alter motor coordination, locomotor and exploratory activities nor induced pro-depressant-like behavior. LOR17, therefore, may emerge as a novel KOPr agonist displaying functional selectivity toward G protein signaling and eliciting antinociceptive/antihypersensitivity effects in different animal models, including oxaliplatin-induced neuropathy.
Collapse
Affiliation(s)
- Andrea Bedini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug and Children Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug and Children Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Monica Baiula
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Gabriela Vaca
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Rossella De Marco
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy.,Department of Agricultural, Food, Enviromental and Animal Science (Di4A), Udine, Italy
| | - Luca Gentilucci
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug and Children Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Santi Spampinato
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
12
|
Schattauer SS, Bedini A, Summers F, Reilly-Treat A, Andrews MM, Land BB, Chavkin C. Reactive oxygen species (ROS) generation is stimulated by κ opioid receptor activation through phosphorylated c-Jun N-terminal kinase and inhibited by p38 mitogen-activated protein kinase (MAPK) activation. J Biol Chem 2019; 294:16884-16896. [PMID: 31575661 PMCID: PMC6851317 DOI: 10.1074/jbc.ra119.009592] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/24/2019] [Indexed: 01/14/2023] Open
Abstract
Activation of the mitogen-activated protein kinase (MAPK) c-Jun N-terminal kinase (JNK) by the Gi/o protein-coupled κ opioid receptor (KOR), μ opioid, and D2 dopamine receptors stimulates peroxiredoxin 6 (PRDX6)-mediated production of reactive oxygen species (ROS). ROS production by KOR-inactivating antagonists norbinaltorphimine (norBNI) and JDTic blocks Gαi protein activation, but the signaling mechanisms and consequences of JNK activation by KOR agonists remain uncharacterized. Binding of arrestins to KOR causes desensitization of G protein signaling and acts as a scaffold to initiate MAPK activation. Here, we found that the KOR agonists U50,488 and dynorphin B stimulated biphasic JNK activation with an early arrestin-independent phase, requiring the small G protein RAC family small GTPase 1 (RAC1) and protein kinase C (PKC), and a later arrestin-scaffolded phase, requiring RAC1 and Ras homolog family member (RHO) kinase. JNK activation by U50,488 and dynorphin B also stimulated PRDX6-dependent ROS production but with an inverted U-shaped dose-response relationship. KOR agonist-induced ROS generation resulted from the early arrestin-independent phase of JNK activation, and this ROS response was suppressed by arrestin-dependent activation of the MAPK p38. The apparent balance between p38 MAPK and JNK/ROS signaling has important physiological implications for understanding of dynorphin activities during the stress response. To visualize these activities, we monitored KOR agonist-mediated activation of ROS in transfected live cells by two fluorescent sensors, CellROX Green and HyPerRed. These findings establish an important aspect of opioid receptor signaling and suggest that ROS induction may be part of the physiological response to KOR activation.
Collapse
Affiliation(s)
- Selena S Schattauer
- Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| | - Andrea Bedini
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Irnerio, 48-40126 Bologna, Italy
| | - Floyd Summers
- Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| | - Aiden Reilly-Treat
- Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| | - Mackenzie M Andrews
- Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
- Department of Bioengineering, University of Washington College of Engineering, Seattle, Washington 98195
| | - Benjamin B Land
- Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| | - Charles Chavkin
- Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| |
Collapse
|
13
|
Dunn AD, Reed B, Erazo J, Ben-Ezra A, Kreek MJ. Signaling Properties of Structurally Diverse Kappa Opioid Receptor Ligands: Toward in Vitro Models of in Vivo Responses. ACS Chem Neurosci 2019; 10:3590-3600. [PMID: 31313902 DOI: 10.1021/acschemneuro.9b00195] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Biased ligands preferentially activate certain signaling pathways downstream of their target receptor, leading to differential physiological or behavioral responses downstream. The kappa opioid receptor (KOR) is a drug target for diseases involving mood and reward, such as depression and addiction. Biased KOR ligands offer the potential to overcome negative side effects that have previously hampered the therapeutic development of KOR agonists by preferentially activating certain signaling pathways. Understanding relationships between ligand bias and behavior is difficult, however, because differences in cellular context and bias quantification methods lead to variation between studies. Here, a set of 21 structurally diverse KOR ligands were tested in parallel, to systematically quantify ligand bias at the KOR. Compounds included the endogenous peptide ligand Dynorphin A(1-17), two novel compounds synthesized for our research, and 18 additional compounds of different structural classes, including morphinans and the natural product Salvinorin A. Compounds were tested for their activity in early KOR signaling pathways (G-protein and β-arrestin recruitment) in KOR-expressing U2OS cells, and ligand bias was calculated. A subset of compounds was tested for sedative properties in the rotarod assay in mice. We found that rotarod sedation significantly correlated with β-arrestin signaling in this system, indicating that this in vitro system can be used to accurately describe this in vivo behavior caused by KOR agonists. Additionally, downstream signaling pathways ERK1/2 and mTOR were evaluated, and we determined that signaling via both of these pathways could diverge from KOR-mediated G-protein and arrestin signaling in this system.
Collapse
Affiliation(s)
- Amelia D. Dunn
- Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Brian Reed
- Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Jose Erazo
- Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Ariel Ben-Ezra
- Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Mary Jeanne Kreek
- Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| |
Collapse
|
14
|
Utility of animal gastrointestinal motility and transit models in functional gastrointestinal disorders. Best Pract Res Clin Gastroenterol 2019; 40-41:101633. [PMID: 31594654 DOI: 10.1016/j.bpg.2019.101633] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/17/2019] [Indexed: 01/31/2023]
Abstract
Alteration in the gastrointestinal (GI) motility and transit comprises an important component of the functional gastrointestinal disorders (FGID). Available animal GI motility and transit models are to study symptoms (delayed gastric emptying, constipation, diarrhea) rather than biological markers to develop an effective treatment that targets the underlying mechanism of altered GI motility in patients. Animal data generated from commonly used methods in human like scintigraphy, breath test and wireless motility capsule may directly translate to the clinic. However, species differences in the control mechanism or pharmacological responses of GI motility may compromise the predictive and translational value of the preclinical data to human. In this review we aim to provide a summary on animal models used to mimic GI motility alteration in FGID, and the impact of the species differences in the physiological and pharmacological responses on the translation of animal GI motility and transit data to human.
Collapse
|
15
|
Li C, Kash TL. κ-Opioid Receptor Modulation of GABAergic Inputs onto Ventrolateral Periaqueductal Gray Dopamine Neurons. MOLECULAR NEUROPSYCHIATRY 2019; 5:190-199. [PMID: 31768372 DOI: 10.1159/000496974] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/15/2019] [Indexed: 11/19/2022]
Abstract
The κ-opioid receptor (KOR) system has been implicated in the regulation of many behaviors including pain. While there are numerous studies suggesting KOR regulation of pain being mediated spinally, there have been reports of pain-like behaviors regulated by central KOR signaling. In particular, oxytocin-induced analgesia appears to be mediated by KOR receptors within the ventrolateral periaqueductal gray (vlPAG). We recently found that activation of dopamine (DA) neurons within the vlPAG is antinociceptive. In this study, we sought to determine the impact of KOR signaling on -GABAergic inputs onto vlPAG DA neurons, and the mechanism through which KOR impacts these inputs. We found that activation of KOR reduced GABAergic transmission onto vlPAG DA neurons. In addition, our data suggest this effect is mediated presynaptically via the G protein βγ-subunit. They raise the possibility that KOR activation disinhibits -vlPAG DA neurons, which could lead to altered regulation of pain-related behaviors.
Collapse
Affiliation(s)
- Chia Li
- Curriculum in Neurobiology, University of North Carolina Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA.,Bowles Center for Alcohol Studies, University of North Carolina Chapel Hill, Chapel Hill, North Carolina, USA
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, University of North Carolina Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Pharmacology, University of North Carolina Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
16
|
Abstract
The nociceptin/orphanin FQ peptide (NOP) receptor-related ligands have been demonstrated in preclinical studies for several therapeutic applications. This article highlights (1) how nonhuman primates (NHP) were used to facilitate the development and application of positron emission tomography tracers in humans; (2) effects of an endogenous NOP ligand, nociceptin/orphanin FQ, and its interaction with mu opioid peptide (MOP) receptor agonists; and (3) promising functional profiles of NOP-related agonists in NHP as analgesics and treatment for substance use disorders. NHP models offer the most phylogenetically appropriate evaluation of opioid and non-opioid receptor functions and drug effects. Based on preclinical and clinical data of ligands with mixed NOP/MOP receptor agonist activity, several factors including their intrinsic efficacies for activating NOP versus MOP receptors and different study endpoints in NHP could contribute to different pharmacological profiles. Ample evidence from NHP studies indicates that bifunctional NOP/MOP receptor agonists have opened an exciting avenue for developing safe, effective medications with fewer side effects for treating pain and drug addiction. In particular, bifunctional NOP/MOP partial agonists hold a great potential as (1) effective spinal analgesics without itch side effects; (2) safe, nonaddictive analgesics without opioid side effects such as respiratory depression; and (3) effective medications for substance use disorders.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
17
|
Estrogen Regulation of GRK2 Inactivates Kappa Opioid Receptor Signaling Mediating Analgesia, But Not Aversion. J Neurosci 2018; 38:8031-8043. [PMID: 30076211 DOI: 10.1523/jneurosci.0653-18.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/11/2018] [Accepted: 07/21/2018] [Indexed: 12/12/2022] Open
Abstract
Activation of κ opioid receptors (KORs) produces analgesia and aversion via distinct intracellular signaling pathways, but whether G protein-biased KOR agonists can be designed to have clinical utility will depend on a better understanding of the signaling mechanisms involved. We found that KOR activation produced conditioned place aversion and potentiated CPP for cocaine in male and female C57BL/6N mice. Consistent with this, males and females both showed arrestin-mediated increases in phospho-p38 MAPK following KOR activation. Unlike in males, however, KOR activation had inconsistent analgesic effects in females and KOR increased Gβγ-mediated ERK phosphorylation in males, but not females. KOR desensitization was not responsible for the lack of response in females because neither Grk3 nor Pdyn gene knock-out enhanced analgesia. Instead, responsiveness was estrous cycle dependent because KOR analgesia was evident during low estrogen phases of the cycle and in ovariectomized (OVX) females. Estradiol treatment of OVX females suppressed KOR-mediated analgesia, demonstrating that estradiol was sufficient to blunt Gβγ-mediated KOR signals. G protein-coupled receptor kinase 2 (GRK2) is known to regulate ERK activation, and we found that the inhibitory, phosphorylated form of GRK2 was significantly higher in intact females. GRK2/3 inhibition by CMPD101 increased KOR stimulation of phospho-ERK in females, decreased sex differences in KOR-mediated inhibition of dopamine release, and enhanced mu opioid receptor and KOR-mediated analgesia in females. In OVX females, estradiol increased the association between GRK2 and Gβγ. These studies suggest that estradiol, through increased phosphorylation of GRK2 and possible sequestration of Gβγ by GRK2, blunts G protein-mediated signals.SIGNIFICANCE STATEMENT Chronic pain disorders are more prevalent in females than males, but opioid receptor agonists show inconsistent analgesic efficacy in females. κ opioid receptor (KOR) agonists have been tested in clinical trials for treating pain disorders based on their analgesic properties and low addictive potential. However, the molecular mechanisms underlying sex differences in KOR actions were previously unknown. Our studies identify an intracellular mechanism involving estradiol regulation of G protein-coupled receptor kinase 2 that is responsible for sexually dimorphic analgesic responses following opioid receptor activation. Understanding this mechanism will be critical for developing effective nonaddictive opioid analgesics for use in women and characterizing sexually dimorphic effects in other inhibitory G protein-coupled receptor signaling responses.
Collapse
|
18
|
Spetea M, Eans SO, Ganno ML, Lantero A, Mairegger M, Toll L, Schmidhammer H, McLaughlin JP. Selective κ receptor partial agonist HS666 produces potent antinociception without inducing aversion after i.c.v. administration in mice. Br J Pharmacol 2017; 174:2444-2456. [PMID: 28494108 PMCID: PMC5513865 DOI: 10.1111/bph.13854] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/09/2017] [Accepted: 05/03/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE The κ receptor has a central role in modulating neurotransmission in central and peripheral neuronal circuits that subserve pain and other behavioural responses. Although κ receptor agonists do not produce euphoria or lead to respiratory suppression, they induce dysphoria and sedation. We hypothesized that brain-penetrant κ receptor ligands possessing biased agonism towards G protein signalling over β-arrestin2 recruitment would produce robust antinociception with fewer associated liabilities. EXPERIMENTAL APPROACH Two new diphenethylamines with high κ receptor selectivity, HS665 and HS666, were assessed following i.c.v. administration in mouse assays of antinociception with the 55°C warm-water tail withdrawal test, locomotor activity in the rotorod and conditioned place preference. The [35 S]-GTPγS binding and β-arrestin2 recruitment in vitro assays were used to characterize biased agonism. KEY RESULTS HS665 (κ receptor agonist) and HS666 (κ receptor partial agonist) demonstrated dose-dependent antinociception after i.c.v. administration mediated by the κ receptor. These highly selective κ receptor ligands displayed varying biased signalling towards G protein coupling in vitro, consistent with a reduced liability profile, reflected by reduced sedation and absence of conditioned place aversion for HS666. CONCLUSIONS AND IMPLICATIONS HS665 and HS666 activate central κ receptors to produce potent antinociception, with HS666 displaying pharmacological characteristics of a κ receptor analgesic with reduced liability for aversive effects correlating with its low efficacy in the β-arrestin2 signalling pathway. Our data provide further understanding of the contribution of central κ receptors in pain suppression, and the prospect of dissociating the antinociceptive effects of HS665 and HS666 from κ receptor-mediated adverse effects.
Collapse
Affiliation(s)
- Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
| | - Shainnel O Eans
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
- Department of PharmacodynamicsUniversity of FloridaGainesvilleFLUSA
| | | | - Aquilino Lantero
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Michael Mairegger
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Lawrence Toll
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
| | - Helmut Schmidhammer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Jay P McLaughlin
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
- Department of PharmacodynamicsUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
19
|
Schattauer SS, Kuhar JR, Song A, Chavkin C. Nalfurafine is a G-protein biased agonist having significantly greater bias at the human than rodent form of the kappa opioid receptor. Cell Signal 2017; 32:59-65. [PMID: 28088389 DOI: 10.1016/j.cellsig.2017.01.016] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/04/2016] [Accepted: 01/09/2017] [Indexed: 11/27/2022]
Abstract
Nalfurafine is a moderately selective kappa opioid receptor (KOR) analgesic with low incidence of dysphoric side effects in clinical development for the treatment of uremic pruritis. The basis for its reduced dysphoric effect compared to other KOR agonists is not clear, but prior studies suggest that the aversive properties of KOR agonists require p38α MAPK activation through an arrestin-dependent mechanism. To determine whether nalfurafine is a functionally selective KOR agonist, we measured its potency to activate the G protein-dependent early phase of Extracellular Signal-Regulated Kinase (ERK1/2) phosphorylation and the arrestin-dependent late phase of p38 MAPK signaling. Nalfurafine was approximately 250 fold more potent for ERK1/2 activation as compared to p38 MAPK activation in human KOR (hKOR) expressing HEK293 cells, and approximately 20 fold more potent for ERK1/2 activation than p38 activation in rodent KOR (rKOR) expressing HEK293 cells. The 10-fold greater G-bias at the hKOR than rKOR was unexpected, however the G protein biased effect of nalfurafine is consistent with its reduced dysphoric effects in human and rodent models. Although nalfurafine is reported to have low receptor selectivity in radioligand binding assays, its antinociceptive effect was blocked by the selective KOR antagonist norbinaltorphimine. Nalfurafine pretreatment also resulted in a KOR-dependent and mu opioid receptor-independent reduction in scratching induced by 5'-GNTI. These findings suggest that nalfurafine is a functionally selective KOR agonist and that KOR agonists able to selectively activate G protein signaling without activating p38α MAPK may have therapeutic potential as non-dysphoric antipruritic analgesics.
Collapse
Affiliation(s)
- Selena S Schattauer
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, United States
| | - Jamie R Kuhar
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, United States
| | - Allisa Song
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, United States
| | - Charles Chavkin
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, United States.
| |
Collapse
|
20
|
Broad J, Maurel D, Kung VWS, Hicks GA, Schemann M, Barnes MR, Kenakin TP, Granier S, Sanger GJ. Human native kappa opioid receptor functions not predicted by recombinant receptors: Implications for drug design. Sci Rep 2016; 6:30797. [PMID: 27492592 PMCID: PMC4974614 DOI: 10.1038/srep30797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/11/2016] [Indexed: 12/14/2022] Open
Abstract
If activation of recombinant G protein-coupled receptors in host cells (by drugs or other ligands) has predictive value, similar data must be obtained with native receptors naturally expressed in tissues. Using mouse and human recombinant κ opioid receptors transfected into a host cell, two selectively-acting compounds (ICI204448, asimadoline) equi-effectively activated both receptors, assessed by measuring two different cell signalling pathways which were equally affected without evidence of bias. In mouse intestine, naturally expressing κ receptors within its nervous system, both compounds also equi-effectively activated the receptor, inhibiting nerve-mediated muscle contraction. However, whereas ICI204448 acted similarly in human intestine, where κ receptors are again expressed within its nervous system, asimadoline was inhibitory only at very high concentrations; instead, low concentrations of asimadoline reduced the activity of ICI204448. This demonstration of species-dependence in activation of native, not recombinant κ receptors may be explained by different mouse/human receptor structures affecting receptor expression and/or interactions with intracellular signalling pathways in native environments, to reveal differences in intrinsic efficacy between receptor agonists. These results have profound implications in drug design for κ and perhaps other receptors, in terms of recombinant-to-native receptor translation, species-dependency and possibly, a need to use human, therapeutically-relevant, not surrogate tissues.
Collapse
Affiliation(s)
- John Broad
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Damien Maurel
- Institut de Génomique Fonctionnelle, Dépt de Pharmacologie Moléculaire, UMR 5203 CNRS-U 661 INSERM, Univ Montpellier I &II, 141, 34094 Montpellier, France
| | - Victor W S Kung
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Gareth A Hicks
- Tioga Pharmaceuticals, 9393 Towne Centre Drive, Suite 200, San Diego, California, USA
| | - Michael Schemann
- Human Biology, TU München, D-85350 Freising-Weihenstephan, Germany
| | - Michael R Barnes
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Terrence P Kenakin
- Dept of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle, Dépt de Pharmacologie Moléculaire, UMR 5203 CNRS-U 661 INSERM, Univ Montpellier I &II, 141, 34094 Montpellier, France
| | - Gareth J Sanger
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| |
Collapse
|
21
|
Zangrandi L, Burtscher J, MacKay JP, Colmers WF, Schwarzer C. The G-protein biased partial κ opioid receptor agonist 6'-GNTI blocks hippocampal paroxysmal discharges without inducing aversion. Br J Pharmacol 2016; 173:1756-67. [PMID: 26928671 PMCID: PMC4867738 DOI: 10.1111/bph.13474] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/05/2016] [Accepted: 02/09/2016] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose With a prevalence of 1–2%, epilepsies belong to the most frequent neurological diseases worldwide. Although antiepileptic drugs are available since several decades, the incidence of patients that are refractory to medication is still over 30%. Antiepileptic effects of κ opioid receptor (κ receptor) agonists have been proposed since the 1980s. However, their clinical use was hampered by dysphoric side effects. Recently, G‐protein biased κ receptor agonists were developed, suggesting reduced aversive effects. Experimental Approach We investigated the effects of the κ receptor agonist U‐50488H and the G‐protein biased partial κ receptor agonist 6′‐GNTI in models of acute seizures and drug‐resistant temporal lobe epilepsy and in the conditioned place avoidance (CPA) test. Moreover, we performed slice electrophysiology to understand the functional mechanisms of 6′‐GNTI. Key Results As previously shown for U‐50488H, 6′‐GNTI markedly increased the threshold for pentylenetetrazole‐induced seizures. All treated mice displayed reduced paroxysmal activity in response to U‐50488H (20 mg·kg−1) or 6′‐GNTI (10–30 nmoles) treatment in the mouse model of intra‐hippocampal injection of kainic acid. Single cell recordings on hippocampal pyramidal cells revealed enhanced inhibitory signalling as potential mechanisms causing the reduction of paroxysmal activity. Effects of 6′‐GNTI were blocked in both seizure models by the κ receptor antagonist 5′‐GNTI. Moreover, 6′‐GNTI did not induce CPA, a measure of aversive effects, while U‐50488H did. Conclusions and Implications Our data provide the proof of principle that anticonvulsant/antiseizure and aversive effects of κ receptor activation can be pharmacologically separated in vivo.
Collapse
Affiliation(s)
- Luca Zangrandi
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Burtscher
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - James P MacKay
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - William F Colmers
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Christoph Schwarzer
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
22
|
Srivastava A, Gupta B, Gupta C, Shukla AK. Emerging Functional Divergence of β-Arrestin Isoforms in GPCR Function. Trends Endocrinol Metab 2015; 26:628-642. [PMID: 26471844 DOI: 10.1016/j.tem.2015.09.001] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/21/2015] [Accepted: 09/02/2015] [Indexed: 12/22/2022]
Abstract
G protein-coupled receptors (GPCRs) are tightly regulated by multifunctional protein β-arrestins. Two isoforms of β-arrestin sharing more than 70% sequence identity and overall very similar 3D structures, β-arrestins 1 and 2, were originally expected to be functionally redundant. However, in recent years multiple lines of emerging evidence suggest they have distinct roles in various aspects of GPCR regulation and signaling. We summarize selected examples of GPCRs where β-arrestin isoforms are discovered to display non-overlapping and sometimes even antagonistic functions. We also discuss potential mechanistic basis for their functional divergence and highlight new frontiers that are likely to form the focal points of research in this area in coming years.
Collapse
Affiliation(s)
- Ashish Srivastava
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Bhagyashri Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Charu Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India.
| |
Collapse
|
23
|
Jamshidi RJ, Jacobs BA, Sullivan LC, Chavera TA, Saylor RM, Prisinzano TE, Clarke WP, Berg KA. Functional selectivity of kappa opioid receptor agonists in peripheral sensory neurons. J Pharmacol Exp Ther 2015; 355:174-82. [PMID: 26297384 PMCID: PMC4613959 DOI: 10.1124/jpet.115.225896] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/19/2015] [Indexed: 01/12/2023] Open
Abstract
Activation of kappa opioid receptors (KORs) expressed by peripheral sensory neurons that respond to noxious stimuli (nociceptors) can reduce neurotransmission of pain stimuli from the periphery to the central nervous system. We have previously shown that the antinociception dose-response curve for peripherally restricted doses of the KOR agonist (-)-(trans)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzeneacetamide (U50488) has an inverted U shape. Here, we found that the downward phase of the U50488 dose-response curve was blocked by an inhibitor of extracellular signal-regulated kinase (ERK) activation U0126. Local administration of the selective KOR agonist salvinorin A (Sal-A), also resulted in an inverted U-shaped curve; however, the downward phase was insensitive to U0126. By contrast, inhibition of c-Jun N-terminal kinase (JNK) partially blocked the downward phase of the dose-response curve to Sal-A, suggesting a role for JNK. In cultures of peripheral sensory neurons, U50488 and Sal-A inhibited adenylyl cyclase activity with similar efficacies; however, their ability to activate ERK and JNK differed. Whereas U50488 activated ERK but not JNK, Sal-A activated JNK but not ERK. Moreover, although both U50488 and Sal-A produced homologous desensitization, desensitization to U50488 was blocked by inhibition of ERK activation, whereas desensitization to Sal-A was blocked by inhibition of JNK. Substitution of an ethoxymethyl ether for the C2 position acetyl group of Sal-A reduced stimulation of JNK, prevented desensitization by ethoxymethyl ether for the C2 position acetyl group of Sal-A, and resulted in a monotonic antinociception dose-response curve. Collectively, these data demonstrate the functional selectivity of KOR ligands for signaling in peripheral sensory neurons, which results in differential effects on behavioral responses in vivo.
Collapse
Affiliation(s)
- Raehannah J Jamshidi
- Department of Pharmacology (R.J.J., B.A.J., L.C.S., T.A.C., W.P.C., K.A.B.), University of Texas Health Science Center, San Antonio, Texas; and Department of Medicinal Chemistry (R.M.S., T.E.P.), University of Kansas School of Pharmacy, Lawrence, Kansas
| | - Blaine A Jacobs
- Department of Pharmacology (R.J.J., B.A.J., L.C.S., T.A.C., W.P.C., K.A.B.), University of Texas Health Science Center, San Antonio, Texas; and Department of Medicinal Chemistry (R.M.S., T.E.P.), University of Kansas School of Pharmacy, Lawrence, Kansas
| | - Laura C Sullivan
- Department of Pharmacology (R.J.J., B.A.J., L.C.S., T.A.C., W.P.C., K.A.B.), University of Texas Health Science Center, San Antonio, Texas; and Department of Medicinal Chemistry (R.M.S., T.E.P.), University of Kansas School of Pharmacy, Lawrence, Kansas
| | - Teresa A Chavera
- Department of Pharmacology (R.J.J., B.A.J., L.C.S., T.A.C., W.P.C., K.A.B.), University of Texas Health Science Center, San Antonio, Texas; and Department of Medicinal Chemistry (R.M.S., T.E.P.), University of Kansas School of Pharmacy, Lawrence, Kansas
| | - Rachel M Saylor
- Department of Pharmacology (R.J.J., B.A.J., L.C.S., T.A.C., W.P.C., K.A.B.), University of Texas Health Science Center, San Antonio, Texas; and Department of Medicinal Chemistry (R.M.S., T.E.P.), University of Kansas School of Pharmacy, Lawrence, Kansas
| | - Thomas E Prisinzano
- Department of Pharmacology (R.J.J., B.A.J., L.C.S., T.A.C., W.P.C., K.A.B.), University of Texas Health Science Center, San Antonio, Texas; and Department of Medicinal Chemistry (R.M.S., T.E.P.), University of Kansas School of Pharmacy, Lawrence, Kansas
| | - William P Clarke
- Department of Pharmacology (R.J.J., B.A.J., L.C.S., T.A.C., W.P.C., K.A.B.), University of Texas Health Science Center, San Antonio, Texas; and Department of Medicinal Chemistry (R.M.S., T.E.P.), University of Kansas School of Pharmacy, Lawrence, Kansas
| | - Kelly A Berg
- Department of Pharmacology (R.J.J., B.A.J., L.C.S., T.A.C., W.P.C., K.A.B.), University of Texas Health Science Center, San Antonio, Texas; and Department of Medicinal Chemistry (R.M.S., T.E.P.), University of Kansas School of Pharmacy, Lawrence, Kansas
| |
Collapse
|
24
|
Al-Saffar A, Nogueira da Costa A, Delaunois A, Leishman DJ, Marks L, Rosseels ML, Valentin JP. Gastrointestinal Safety Pharmacology in Drug Discovery and Development. Handb Exp Pharmacol 2015; 229:291-321. [PMID: 26091645 DOI: 10.1007/978-3-662-46943-9_12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although the basic structure of the gastrointestinal tract (GIT) is similar across species, there are significant differences in the anatomy, physiology, and biochemistry between humans and laboratory animals, which should be taken into account when conducting a gastrointestinal (GI) assessment. Historically, the percentage of cases of drug attrition associated with GI-related adverse effects is small; however, this incidence has increased over the last few years. Drug-related GI effects are very diverse, usually functional in nature, and not limited to a single pharmacological class. The most common GI signs are nausea and vomiting, diarrhea, constipation, and gastric ulceration. Despite being generally not life-threatening, they can greatly affect patient compliance and quality of life. There is therefore a real need for improved and/or more extensive GI screening of candidate drugs in preclinical development, which may help to better predict clinical effects. Models to identify drug effects on GI function cover GI motility, nausea and emesis liability, secretory function (mainly gastric secretion), and absorption aspects. Both in vitro and in vivo assessments are described in this chapter. Drug-induced effects on GI function can be assessed in stand-alone safety pharmacology studies or as endpoints integrated into toxicology studies. In silico approaches are also being developed, such as the gut-on-a-chip model, but await further optimization and validation before routine use in drug development. GI injuries are still in their infancy with regard to biomarkers, probably due to their greater diversity. Nevertheless, several potential blood, stool, and breath biomarkers have been investigated. However, additional validation studies are necessary to assess the relevance of these biomarkers and their predictive value for GI injuries.
Collapse
Affiliation(s)
- Ahmad Al-Saffar
- Faculty of Medicine, Department of Medical Sciences, Uppsala University, 751 85, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
25
|
Dogra S, Yadav PN. Biased agonism at kappa opioid receptors: Implication in pain and mood disorders. Eur J Pharmacol 2015; 763:184-90. [PMID: 26164787 DOI: 10.1016/j.ejphar.2015.07.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 06/18/2015] [Accepted: 07/07/2015] [Indexed: 10/23/2022]
Abstract
The kappa opioid receptor (k receptor) and its endogenous ligand dynorphin have received significant attention due to their involvement in pathophysiology of mood disorders, drug addiction, psychotic disorders and pain. Multiple lines of evidences suggest that the k receptor modulates overlapping neurocircuits connecting brainstem monoaminergic nuclei with forebrain limbic structures and thereby regulates neurobiological effects of stress and psychostimulants. The emerging concept of "biased agonism" (also known as functional selectivity) for G Protein Coupled Receptor (GPCR) ligands have provided new insights into overall response generated by a ligand, which could be exploited for drug discovery. According to this concept, every ligand possesses the unique ability (coded in its structure) that dictates distinct signalling pattern, and consequently beneficial or adverse response. Although still a long way to comprehend the clinical potential of biased GPCR ligands, such ligand could be vital pharmacological probes. This article highlights various lines of evidence, which indicates different ligands of k receptor as "biased", and their potential implications in mood and pain disorders.
Collapse
Affiliation(s)
- Shalini Dogra
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Prem N Yadav
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India.
| |
Collapse
|
26
|
DiMattio KM, Ehlert FJ, Liu-Chen LY. Intrinsic relative activities of κ opioid agonists in activating Gα proteins and internalizing receptor: Differences between human and mouse receptors. Eur J Pharmacol 2015; 761:235-44. [PMID: 26057692 DOI: 10.1016/j.ejphar.2015.05.054] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/17/2015] [Accepted: 05/18/2015] [Indexed: 12/21/2022]
Abstract
Several investigators recently identified biased κ opioid receptor (KOP receptor) agonists. However, no comprehensive study of the functional selectivity of available KOP receptor agonists at the human and mouse KOP receptors (hKOP receptor and mKOP receptor, respectively) has been published. Here we examined the ability of over 20 KOP receptor agonists to activate G proteins and to internalize the receptor. Clonal neuro-2a mouse neuroblastoma (N2a) cells stably transfected with the hKOP receptor or mKOP receptor were used. We employed agonist-induced [(35)S]GTPγS binding and KOP receptor internalization as measures of activation of G protein and β-arrestin pathways, respectively. The method of Ehlert and colleagues was used to quantify intrinsic relative activities at G protein activation (RAi-G) and receptor internalization (RAi-I) and the degree of functional selectivity between the two [Log RAi-G - logRAi-I, RAi-G/RAi-I and bias factor]. The parameter, RAi, represents a relative estimate of agonist affinity for the active receptor state that elicits a given response. The endogenous ligand dynorphin A (1-17) was designated as the balanced ligand with a bias factor of 1. Interestingly, we found that there were species differences in functional selectivity. The most striking differences were for 12-epi-salvinorin A, U69,593, and ICI-199,441. 12-Epi-salvinorin A was highly internalization-biased at the mKOP receptor, but apparently G protein-biased at hKOP receptor. U69,593 was much more internalization-biased at mKOP receptor than hKOP receptor. ICI199,441 showed internalization-biased at the mKOP receptor and G protein-biased at the hKOP receptor. Possible mechanisms for the observed species differences are discussed.
Collapse
Affiliation(s)
- Kelly M DiMattio
- Center for Substance Abuse Research and Department of Pharmacology, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA.
| | - Frederick J Ehlert
- Department of Pharmacology, School of Medicine, University of California, Irvine, CA 92697, USA.
| | - Lee-Yuan Liu-Chen
- Center for Substance Abuse Research and Department of Pharmacology, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
27
|
Filipeanu CM, Pullikuth AK, Guidry JJ. Molecular determinants of the human α2C-adrenergic receptor temperature-sensitive intracellular traffic. Mol Pharmacol 2015; 87:792-802. [PMID: 25680754 PMCID: PMC4407737 DOI: 10.1124/mol.114.096198] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/12/2015] [Indexed: 01/22/2023] Open
Abstract
The human α2C-adrenergic receptor (α2C-AR) is localized intracellularly at physiologic temperature. Decreasing the environmental temperature strongly stimulates the receptor transport to the cell surface. In contrast, rat and mouse α2C-AR plasma membrane levels are less sensitive to decrease in temperature, whereas the opossum α2C-AR cell surface levels are not changed in these conditions. Structural analysis demonstrated that human α2C-AR has a high number of arginine residues in the third intracellular loop and in the C-terminus, organized as putative RXR motifs. Although these motifs do not affect the receptor subcellular localization at 37°C, deletion of the arginine clusters significantly enhanced receptor plasma membrane levels at reduced temperature. We found that this exaggerated transport of the human receptor is mediated by two functional arginine clusters, one in the third intracellular loop and one in the C-terminus. This effect is mediated by interactions with COPI vesicles, but not by 14-3-3 proteins. In rat α2C-AR, the arginine cluster from the third intracellular loop is shifted to the left due to three missing residues. Reinsertion of these residues in the rat α2C-AR restored the same temperature sensitivity as in the human receptor. Proteomic and coimmunoprecipitation experiments identified pontin as a molecule having stronger interactions with human α2C-AR compared with rat α2C-AR. Inhibition of pontin activity enhanced human receptor plasma membrane levels and signaling at 37°C. Our results demonstrate that human α2C-AR has a unique temperature-sensitive traffic pattern within the G protein-coupled receptor class due to interactions with different molecular chaperones, mediated in part by strict spatial localization of specific arginine residues.
Collapse
Affiliation(s)
- Catalin M Filipeanu
- Department of Pharmacology, College of Medicine, Howard University, Washington, DC (C.M.F.); Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana (A.K.P., J.J.G.); Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina (A.K.P.); and Louisiana State University Health Sciences Center Proteomics Core Facility, New Orleans, Louisiana (J.J.G.)
| | - Ashok K Pullikuth
- Department of Pharmacology, College of Medicine, Howard University, Washington, DC (C.M.F.); Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana (A.K.P., J.J.G.); Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina (A.K.P.); and Louisiana State University Health Sciences Center Proteomics Core Facility, New Orleans, Louisiana (J.J.G.)
| | - Jessie J Guidry
- Department of Pharmacology, College of Medicine, Howard University, Washington, DC (C.M.F.); Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana (A.K.P., J.J.G.); Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina (A.K.P.); and Louisiana State University Health Sciences Center Proteomics Core Facility, New Orleans, Louisiana (J.J.G.)
| |
Collapse
|
28
|
White KL, Robinson JE, Zhu H, DiBerto JF, Polepally PR, Zjawiony JK, Nichols DE, Malanga CJ, Roth BL. The G protein-biased κ-opioid receptor agonist RB-64 is analgesic with a unique spectrum of activities in vivo. J Pharmacol Exp Ther 2015; 352:98-109. [PMID: 25320048 PMCID: PMC4279099 DOI: 10.1124/jpet.114.216820] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 10/10/2014] [Indexed: 01/07/2023] Open
Abstract
The hypothesis that functionally selective G protein-coupled receptor (GPCR) agonists may have enhanced therapeutic benefits has revitalized interest for many GPCR targets. In particular, although κ-opioid receptor (KOR) agonists are analgesic with a low risk of dependence and abuse, their use is limited by a propensity to induce sedation, motor incoordination, hallucinations, and dysphoria-like states. Several laboratories have produced a body of work suggesting that G protein-biased KOR agonists might be analgesic with fewer side effects. Although that has been an intriguing hypothesis, suitable KOR-selective and G protein-biased agonists have not been available to test this idea. Here we provide data using a G protein-biased agonist, RB-64 (22-thiocyanatosalvinorin A), which suggests that KOR-mediated G protein signaling induces analgesia and aversion, whereas β-arrestin-2 signaling may be associated with motor incoordination. Additionally, unlike unbiased KOR agonists, the G protein-biased ligand RB-64 does not induce sedation and does not have anhedonia-like actions, suggesting that a mechanism other than G protein signaling mediates these effects. Our findings provide the first evidence for a highly selective and G protein-biased tool compound for which many, but not all, of the negative side effects of KOR agonists can be minimized by creating G protein-biased KOR agonists.
Collapse
Affiliation(s)
- Kate L White
- Department of Pharmacology (K.L.W., H.Z., D.E.N., B.L.R.), Department of Neurology (J.E.R., J.F.D., C.J.M.), and Bowles Center for Alcohol Studies (J.E.R., C.J.M.), University of North Carolina, Chapel Hill, North Carolina; and Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, Mississippi (P.R.P., J.K.Z.)
| | - J Elliott Robinson
- Department of Pharmacology (K.L.W., H.Z., D.E.N., B.L.R.), Department of Neurology (J.E.R., J.F.D., C.J.M.), and Bowles Center for Alcohol Studies (J.E.R., C.J.M.), University of North Carolina, Chapel Hill, North Carolina; and Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, Mississippi (P.R.P., J.K.Z.)
| | - Hu Zhu
- Department of Pharmacology (K.L.W., H.Z., D.E.N., B.L.R.), Department of Neurology (J.E.R., J.F.D., C.J.M.), and Bowles Center for Alcohol Studies (J.E.R., C.J.M.), University of North Carolina, Chapel Hill, North Carolina; and Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, Mississippi (P.R.P., J.K.Z.)
| | - Jeffrey F DiBerto
- Department of Pharmacology (K.L.W., H.Z., D.E.N., B.L.R.), Department of Neurology (J.E.R., J.F.D., C.J.M.), and Bowles Center for Alcohol Studies (J.E.R., C.J.M.), University of North Carolina, Chapel Hill, North Carolina; and Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, Mississippi (P.R.P., J.K.Z.)
| | - Prabhakar R Polepally
- Department of Pharmacology (K.L.W., H.Z., D.E.N., B.L.R.), Department of Neurology (J.E.R., J.F.D., C.J.M.), and Bowles Center for Alcohol Studies (J.E.R., C.J.M.), University of North Carolina, Chapel Hill, North Carolina; and Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, Mississippi (P.R.P., J.K.Z.)
| | - Jordan K Zjawiony
- Department of Pharmacology (K.L.W., H.Z., D.E.N., B.L.R.), Department of Neurology (J.E.R., J.F.D., C.J.M.), and Bowles Center for Alcohol Studies (J.E.R., C.J.M.), University of North Carolina, Chapel Hill, North Carolina; and Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, Mississippi (P.R.P., J.K.Z.)
| | - David E Nichols
- Department of Pharmacology (K.L.W., H.Z., D.E.N., B.L.R.), Department of Neurology (J.E.R., J.F.D., C.J.M.), and Bowles Center for Alcohol Studies (J.E.R., C.J.M.), University of North Carolina, Chapel Hill, North Carolina; and Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, Mississippi (P.R.P., J.K.Z.)
| | - C J Malanga
- Department of Pharmacology (K.L.W., H.Z., D.E.N., B.L.R.), Department of Neurology (J.E.R., J.F.D., C.J.M.), and Bowles Center for Alcohol Studies (J.E.R., C.J.M.), University of North Carolina, Chapel Hill, North Carolina; and Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, Mississippi (P.R.P., J.K.Z.)
| | - Bryan L Roth
- Department of Pharmacology (K.L.W., H.Z., D.E.N., B.L.R.), Department of Neurology (J.E.R., J.F.D., C.J.M.), and Bowles Center for Alcohol Studies (J.E.R., C.J.M.), University of North Carolina, Chapel Hill, North Carolina; and Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, Mississippi (P.R.P., J.K.Z.)
| |
Collapse
|
29
|
Rutten K, Tzschentke TM, Koch T, Schiene K, Christoph T. Pharmacogenomic study of the role of the nociceptin/orphanin FQ receptor and opioid receptors in diabetic hyperalgesia. Eur J Pharmacol 2014; 741:264-71. [PMID: 25169429 DOI: 10.1016/j.ejphar.2014.08.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 07/10/2014] [Accepted: 08/05/2014] [Indexed: 01/03/2023]
Abstract
Targeting functionally independent receptors may provide synergistic analgesic effects in neuropathic pain. To examine the interdependency between different opioid receptors (µ-opioid peptide [MOP], δ-opioid peptide [DOP] and κ-opioid peptide [KOP]) and the nociceptin/orphanin FQ peptide (NOP) receptor in streptozotocin (STZ)-induced diabetic polyneuropathy, nocifensive activity was measured using a hot plate test in wild-type and NOP, MOP, DOP and KOP receptor knockout mice in response to the selective receptor agonists Ro65-6570, morphine, SNC-80 and U50488H, or vehicle. Nocifensive activity was similar in non-diabetic wild-type and knockout mice at baseline, before agonist or vehicle administration. STZ-induced diabetes significantly increased heat sensitivity in all mouse strains, but MOP, DOP and KOP receptor knockouts showed a smaller degree of hyperalgesia than wild-type mice and NOP receptor knockouts. For each agonist, a significant antihyperalgesic effect was observed in wild-type diabetic mice (all P<0.05 versus vehicle); the effect was markedly attenuated in diabetic mice lacking the cognate receptor compared with wild-type diabetic mice. Morphine was the only agonist that demonstrated near-full antihyperalgesic efficacy across all non-cognate receptor knockouts. Partial or near-complete reductions in efficacy were observed with Ro65-6570 in DOP and KOP receptor knockouts, with SNC-80 in NOP, MOP and KOP receptor knockouts, and with U50488H in NOP and DOP receptor knockouts. There was no evidence of NOP and MOP receptor interdependency in response to selective agonists for these receptors. These findings suggest that concurrent activation of NOP and MOP receptors, which showed functional independence, may yield an effective and favorable therapeutic analgesic profile.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Analgesics, Opioid/therapeutic use
- Animals
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Hyperalgesia/drug therapy
- Hyperalgesia/genetics
- Hyperalgesia/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid/physiology
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/physiology
- Nociceptin Receptor
Collapse
Affiliation(s)
- Kris Rutten
- Grünenthal GmbH, Global Preclinical Drug Development, Department of Pain Pharmacology, Zieglerstrasse 6, 52078 Aachen, Germany.
| | - Thomas M Tzschentke
- Grünenthal GmbH, Global Preclinical Drug Development, Department of Pain Pharmacology, Zieglerstrasse 6, 52078 Aachen, Germany
| | - Thomas Koch
- Grünenthal GmbH, Global Preclinical Drug Development, Department of Molecular Pharmacology, Aachen, Germany
| | - Klaus Schiene
- Grünenthal GmbH, Global Preclinical Drug Development, Department of Pain Pharmacology, Zieglerstrasse 6, 52078 Aachen, Germany
| | - Thomas Christoph
- Grünenthal GmbH, Global Preclinical Drug Development, Department of Pain Pharmacology, Zieglerstrasse 6, 52078 Aachen, Germany
| |
Collapse
|
30
|
Broad J, Góralczyk A, Mannur K, Dukes GE, Sanger GJ. Drugs acting at 5-HT4 , D2 , motilin, and ghrelin receptors differ markedly in how they affect neuromuscular functions in human isolated stomach. Neurogastroenterol Motil 2014; 26:851-61. [PMID: 24750304 DOI: 10.1111/nmo.12338] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/10/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Progress in identifying safer, effective drugs to increase gastric emptying is impeded by failed clinical trials. One potential reason for failure is lack of translation from animal models to the human condition. To make progress, the actions of existing drugs and new therapeutic candidates need to be understood in human isolated stomach. METHODS Neuromuscular activities were evoked in human gastric antrum circular muscle by electrical field stimulation (EFS), defined phenotypically using pharmacological tools. KEY RESULTS EFS evoked cholinergically mediated contractions, attenuated by simultaneous nitrergic activation. The 5-HT4 receptor agonist/D2 antagonist metoclopramide and the selective 5-HT4 agonist prucalopride, facilitated contractions in the absence (respectively, Emax 95 ± 29% and 42 ± 9%, n = 3-6 each concentration) and presence (139 ± 38%, 55 ± 13%, n = 3-5) of the NO synthase inhibitor L-NAME, without affecting submaximal contractions to carbachol; the 5-HT4 antagonist SB204070 prevented facilitation by metoclopramide 100 μM (respectively, -5 (range -26 to 34) and 167 (12-1327)% in presence and absence; n = 5-6). The selective motilin receptor agonist camicinal provided considerably greater facilitation (478 (12-2080)% at 30 μM, n = 8). Domperidone (0.001-100 μM; n = 3-6) and acylated or des-acylated ghrelin (1-300 nM; n = 2-4) had no consistent activity, even with protease inhibitors. CONCLUSIONS & INFERENCES 5-HT4 receptor agonists show different efficacies. Motilin receptor activation has greater potential to increase gastric emptying, whereas ghrelin and D2 receptor antagonism have no direct activity. Drugs stimulating human gastric motility directly can act regardless of disease mechanisms, whereas drugs without direct activity but an ability to block nausea/vomiting may be effective only if these symptoms exist.
Collapse
Affiliation(s)
- J Broad
- Neurogastroenterology Group, Blizard Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | | | | |
Collapse
|
31
|
Gimenez LE, Vishnivetskiy SA, Gurevich VV. Targeting individual GPCRs with redesigned nonvisual arrestins. Handb Exp Pharmacol 2014; 219:153-70. [PMID: 24292829 DOI: 10.1007/978-3-642-41199-1_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Numerous human diseases are caused by excessive signaling of mutant G protein-coupled receptors (GPCRs) or receptors that are overstimulated due to upstream signaling imbalances. The feasibility of functional compensation by arrestins with enhanced ability to quench receptor signaling was recently tested in the visual system. The results showed that even in this extremely demanding situation of rods that have no ability to phosphorylate rhodopsin, enhanced arrestin improved rod morphology, light sensitivity, survival, and accelerated photoresponse recovery. Structurally distinct enhanced mutants of arrestins that bind phosphorylated and non-phosphorylated active GPCRs with much higher affinity than parental wild-type (WT) proteins have been constructed. These "super-arrestins" are likely to have the power to dampen the signaling by hyperactive GPCRs. However, most cells express 5-20 GPCR subtypes, only one of which would be overactive, while nonvisual arrestins are remarkably promiscuous, binding hundreds of different GPCRs. Thus, to be therapeutically useful, enhanced versions of nonvisual arrestins must be made fairly specific for particular receptors. Recent identification of very few arrestin residues as key receptor discriminators paves the way to the construction of receptor subtype-specific nonvisual arrestins.
Collapse
Affiliation(s)
- Luis E Gimenez
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, Nashville, TN, 37232, USA,
| | | | | |
Collapse
|
32
|
Bidlack JM. Mixed κ/μ partial opioid agonists as potential treatments for cocaine dependence. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:387-418. [PMID: 24484983 DOI: 10.1016/b978-0-12-420118-7.00010-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cocaine use activates the dopamine reward pathway, leading to the reinforcing effects of dopamine. There is no FDA-approved medication for treating cocaine dependence. Opioid agonists and antagonists have been approved for treating opioid and alcohol dependence. Agonists that activate the μ opioid receptor increase dopamine levels in the nucleus accumbens, while μ receptor antagonists decrease dopamine levels by blocking the effects of endogenous opioid peptides. Activation of the κ opioid receptor decreases dopamine levels and leads to dysphoria. In contrast, inhibition of the κ opioid receptor decreases dopamine levels in the nucleus accumbens. Antagonists acting at the κ receptor reduce stress-mediated behaviors and anxiety. Mixed partial μ/κ agonists have the potential of striking a balance between dopamine levels and attenuating relapse to cocaine. The pharmacological properties of mixed μ/κ opioid receptor agonists will be discussed and results from clinical and preclinical studies will be presented. Results from studies with some of the classical benzomorphans and morphinans will be presented as they lay the foundation for structure-activity relationships. Recent results with other partial opioid agonists, including buprenorphine derivatives and the mixed μ/κ peptide CJ-15,208, will be discussed. The behavioral effects of the mixed μ/κ MCL-741, an aminothiazolomorphinan, in attenuating cocaine-induced locomotor activity will be presented. While not a mixed μ/κ opioid, results obtained with GSK1521498, a μ receptor inverse agonist, will be discussed. Preclinical strategies and successes will lay the groundwork for the further development of mixed μ/κ opioid receptor agonists to treat cocaine dependence.
Collapse
Affiliation(s)
- Jean M Bidlack
- School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA.
| |
Collapse
|
33
|
Lalanne L, Ayranci G, Kieffer BL, Lutz PE. The kappa opioid receptor: from addiction to depression, and back. Front Psychiatry 2014; 5:170. [PMID: 25538632 PMCID: PMC4258993 DOI: 10.3389/fpsyt.2014.00170] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 11/13/2014] [Indexed: 12/16/2022] Open
Abstract
Comorbidity is a major issue in psychiatry that notably associates with more severe symptoms, longer illness duration, and higher service utilization. Therefore, identifying key clusters of comorbidity and exploring the underlying pathophysiological mechanisms represent important steps toward improving mental health care. In the present review, we focus on the frequent association between addiction and depression. In particular, we summarize the large body of evidence from preclinical models indicating that the kappa opioid receptor (KOR), a member of the opioid neuromodulatory system, represents a central player in the regulation of both reward and mood processes. Current data suggest that the KOR modulates overlapping neuronal networks linking brainstem monoaminergic nuclei with forebrain limbic structures. Rewarding properties of both drugs of abuse and natural stimuli, as well as the neurobiological effects of stressful experiences, strongly interact at the level of KOR signaling. In addiction models, activity of the KOR is potentiated by stressors and critically controls drug-seeking and relapse. In depression paradigms, KOR signaling is responsive to a variety of stressors, and mediates despair-like responses. Altogether, the KOR represents a prototypical substrate of comorbidity, whereby life experiences converge upon common brain mechanisms to trigger behavioral dysregulation and increased risk for distinct but interacting psychopathologies.
Collapse
Affiliation(s)
- Laurence Lalanne
- CNRS UMR-7104, Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U-964, Université de Strasbourg , Illkirch , France ; Department of Psychiatry, University Hospital of Strasbourg and Medical School of Strasbourg , Strasbourg , France
| | - Gulebru Ayranci
- CNRS UMR-7104, Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U-964, Université de Strasbourg , Illkirch , France ; Douglas Mental Health Institute, McGill University , Montréal, QC , Canada
| | - Brigitte L Kieffer
- Douglas Mental Health Institute, McGill University , Montréal, QC , Canada
| | - Pierre-Eric Lutz
- Douglas Mental Health Institute, McGill University , Montréal, QC , Canada
| |
Collapse
|
34
|
Arrestin-mediated activation of p38 MAPK: molecular mechanisms and behavioral consequences. Handb Exp Pharmacol 2014; 219:281-92. [PMID: 24292835 DOI: 10.1007/978-3-642-41199-1_14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Studies of kappa opioid receptor signaling mechanisms during the last decade have demonstrated that agonist activation of the receptor results in Gβγ-dependent signaling and distinct arrestin-dependent signaling events. Gβγ-dependent signaling results in ion channel regulation causing neuronal inhibition, inhibition of transmitter release, and subsequent analgesic responses. In contrast, arrestin-dependent signaling events result in p38 MAPK activation and subsequent dysphoric and proaddictive behavioral responses. Resolution of these two branches of signaling cascades has enabled strategies designed to identify pathway-selective drugs that may have unique therapeutic utilities.
Collapse
|
35
|
Abstract
This paper is the thirty-fifth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2012 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 (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
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
|
36
|
Zhan X, Kaoud TS, Kook S, Dalby KN, Gurevich VV. JNK3 enzyme binding to arrestin-3 differentially affects the recruitment of upstream mitogen-activated protein (MAP) kinase kinases. J Biol Chem 2013; 288:28535-28547. [PMID: 23960075 PMCID: PMC3789954 DOI: 10.1074/jbc.m113.508085] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Indexed: 12/29/2022] Open
Abstract
Arrestin-3 was previously shown to bind JNK3α2, MKK4, and ASK1. However, full JNK3α2 activation requires phosphorylation by both MKK4 and MKK7. Using purified proteins we show that arrestin-3 directly interacts with MKK7 and promotes JNK3α2 phosphorylation by both MKK4 and MKK7 in vitro as well as in intact cells. The binding of JNK3α2 promotes an arrestin-3 interaction with MKK4 while reducing its binding to MKK7. Interestingly, the arrestin-3 concentration optimal for scaffolding the MKK7-JNK3α2 module is ∼10-fold higher than for the MKK4-JNK3α2 module. The data provide a mechanistic basis for arrestin-3-dependent activation of JNK3α2. The opposite effects of JNK3α2 on arrestin-3 interactions with MKK4 and MKK7 is the first demonstration that the kinase components in mammalian MAPK cascades regulate each other's interactions with a scaffold protein. The results show how signaling outcomes can be affected by the relative expression of scaffolding proteins and components of signaling cascades that they assemble.
Collapse
Affiliation(s)
- Xuanzhi Zhan
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232 and
| | - Tamer S. Kaoud
- Division of Medicinal Chemistry, The University of Texas at Austin, Austin, Texas 78712
| | - Seunghyi Kook
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232 and
| | - Kevin N. Dalby
- Division of Medicinal Chemistry, The University of Texas at Austin, Austin, Texas 78712
| | - Vsevolod V. Gurevich
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232 and
| |
Collapse
|
37
|
Abstract
κ opioid receptors (KORs) belong to the G-protein-coupled class of receptors (GPCRs). They are activated by the endogenous opioid peptide dynorphin (DYN) and expressed at particularly high levels within brain areas implicated in modulation of motivation, emotion, and cognitive function. Chronic activation of KORs in animal models has maladaptive effects including increases in behaviors that reflect depression, the propensity to engage in drug-seeking behavior, and drug craving. The fact that KOR activation has such a profound influence on behaviors often triggered by stress has led to interest in selective KOR antagonists as potential therapeutic agents. This Perspective provides a description of preclinical research conducted in the development of several different classes of selective KOR antagonists, a summary of the clinical studies conducted thus far, and recommendations for the type of work needed in the future to determine if these agents would be useful as pharmacotherapies for neuropsychiatric illness.
Collapse
Affiliation(s)
- F Ivy Carroll
- Center for Organic and Medicinal Chemistry, Research Triangle Institute , P.O. Box 12194, Research Triangle Park, North Carolina 27709, USA.
| | | |
Collapse
|
38
|
Hirsch S, Corradini L, Just S, Arndt K, Doods H. The CGRP receptor antagonist BIBN4096BS peripherally alleviates inflammatory pain in rats. Pain 2013; 154:700-707. [PMID: 23473785 DOI: 10.1016/j.pain.2013.01.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 12/21/2012] [Accepted: 01/05/2013] [Indexed: 11/30/2022]
Abstract
Calcitonin gene-related peptide (CGRP) is known to play a major role in the pathogenesis of pain syndromes, in particular migraine pain. Here we focus on its implication in a rat pain model of inflammation, induced by injection of complete Freund adjuvant (CFA). The nonpeptide CGRP receptor antagonist BIBN4096BS reduces migraine pain and trigeminal neuronal activity. Here we demonstrate that the compound reduces inflammatory pain and spinal neuronal activity. Behavioural experiments reveal a reversal of the CFA-induced mechanical hypersensitivity and monoiodoacetate (MIA)-induced weight-bearing deficit in rats after systemic drug administration. To further investigate the mechanism of action of the CGRP antagonist in inflammatory pain, in vivo electrophysiological studies were performed in CFA-injected rats. Recordings from wide dynamic range neurons in deep dorsal horn layers of the lumbar spinal cord confirmed a reduction of neuronal activity after systemic drug application. The same amount of reduction occurred after topical administration onto the paw, with resulting systemic plasma concentrations in the low nanomolar range. However, spinal administration of BIBN4096BS did not modify the neuronal activity in the CFA model. Peripheral blockade of CGRP receptors by BIBN4096BS significantly alleviates inflammatory pain.
Collapse
Affiliation(s)
- Silke Hirsch
- Department of CNS Diseases Research, Birkendorfer Strasse 65, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany
| | | | | | | | | |
Collapse
|