1
|
De Neve J, Breault É, Previti S, Vangeloven E, Loranger B, Chartier M, Brouillette R, Lanoie A, Holleran BJ, Longpré JM, Gendron L, Tourwé D, Sarret P, Ballet S. Design, Synthesis, and In Vitro Characterization of Proteolytically-Stable Opioid-Neurotensin Hybrid Peptidomimetics. ACS Pharmacol Transl Sci 2024; 7:2784-2798. [PMID: 39296263 PMCID: PMC11406707 DOI: 10.1021/acsptsci.4c00236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 09/21/2024]
Abstract
Linking an opioid to a nonopioid pharmacophore represents a promising approach for reducing opioid-induced side effects during pain management. Herein, we describe the optimization of the previously reported opioid-neurotensin hybrids (OPNT-hybrids), SBL-OPNT-05 & -10, containing the μ-/δ-opioid agonist H-Dmt-d-Arg-Aba-β-Ala-NH2 and NT(8-13) analogs optimized for NTS2 affinity. In the present work, the constrained dipeptide Aba-β-Ala was modified to investigate the optimal linker length between the two pharmacophores, as well as the effect of expanding the aromatic moiety within constrained dipeptide analogs, via the inclusion of a naphthyl moiety. Additionally, the N-terminal Arg residue of the NT(8-13) pharmacophore was substituted with β3 hArg. For all analogs, affinity was determined at the MOP, DOP, NTS1, and NTS2 receptors. Several of the hybrid ligands showed a subnanomolar affinity for MOP, improved binding for DOP compared to SBL-OPNT-05 & -10, as well as an excellent NTS2-affinity with high selectivity over NTS1. Subsequently, the Gαi1 and β-arrestin-2 pathways were evaluated for all hybrids, along with their stability in rat plasma. Upon MOP activation, SBL-OPNT-13 and -18 were the least effective at recruiting β-arrestin-2 (E max = 17 and 12%, respectively), while both compounds were also found to be partial agonists at the Gαi1 pathway, despite improved potency compared to DAMGO. Importantly, these analogs also showed a half-life in rat plasma in excess of 48 h, making them valuable tools for future in vivo investigations.
Collapse
Affiliation(s)
- Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Émile Breault
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Santo Previti
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Esaü Vangeloven
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Bobbi Loranger
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Magali Chartier
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Rebecca Brouillette
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Annik Lanoie
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Brian J Holleran
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Jean-Michel Longpré
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Louis Gendron
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Philippe Sarret
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| |
Collapse
|
2
|
Puls K, Olivé-Marti AL, Hongnak S, Lamp D, Spetea M, Wolber G. Discovery of Novel, Selective, and Nonbasic Agonists for the Kappa-Opioid Receptor Determined by Salvinorin A-Based Virtual Screening. J Med Chem 2024; 67:13788-13801. [PMID: 39088801 PMCID: PMC11345774 DOI: 10.1021/acs.jmedchem.4c00590] [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: 03/11/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 08/03/2024]
Abstract
Modulating the kappa-opioid receptor (KOR) is a promising strategy for treating various human diseases. KOR agonists show potential for treating pain, pruritus, and epilepsy, while KOR antagonists show potential for treating depression, anxiety, and addiction. The diterpenoid Salvinorin A (SalA), a secondary metabolite of Salvia divinorum, is a potent and selective KOR agonist. Unlike typical opioids, SalA lacks a basic nitrogen, which encouraged us to search for nonbasic KOR ligands. Through structure-based virtual screening using 3D pharmacophore models based on the binding mode of SalA, we identified novel, nonbasic, potent, and selective KOR agonists. In vitro studies confirmed two virtual hits, SalA-VS-07 and SalA-VS-08, as highly selective for the KOR and showing G protein-biased KOR agonist activity. Both KOR ligands share a novel spiro-moiety and a nonbasic scaffold. Our findings provide novel starting points for developing therapeutics aimed at treating pain and other conditions in which KOR is a central player.
Collapse
Affiliation(s)
- Kristina Puls
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| | - Aina-Leonor Olivé-Marti
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Siriwat Hongnak
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - David Lamp
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Mariana Spetea
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Gerhard Wolber
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| |
Collapse
|
3
|
De Neve J, Elhabazi K, Gonzalez S, Herby C, Schneider S, Utard V, Fellmann-Clauss R, Petit-Demouliere N, Lecat S, Kremer M, Ces A, Daubeuf F, Martin C, Ballet S, Bihel F, Simonin F. Multitarget μ-Opioid Receptor Agonists─Neuropeptide FF Receptor Antagonists Induce Potent Antinociception with Reduced Adverse Side Effects. J Med Chem 2024; 67:7603-7619. [PMID: 38687204 DOI: 10.1021/acs.jmedchem.4c00442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The design of bifunctional compounds is a promising approach toward the development of strong analgesics with reduced side effects. We here report the optimization of the previously published lead peptide KGFF09, which contains opioid receptor agonist and neuropeptide FF receptor antagonist pharmacophores and is shown to induce potent antinociception and reduced side effects. We evaluated the novel hybrid peptides for their in vitro activity at MOP, NPFFR1, and NPFFR2 and selected four of them (DP08/14/32/50) for assessment of their acute antinociceptive activity in mice. We further selected DP32 and DP50 and observed that their antinociceptive activity is mostly peripherally mediated; they produced no respiratory depression, no hyperalgesia, significantly less tolerance, and strongly attenuated withdrawal syndrome, as compared to morphine and the recently FDA-approved TRV130. Overall, these data suggest that MOP agonist/NPFF receptor antagonist hybrids might represent an interesting strategy to develop novel analgesics with reduced side effects.
Collapse
MESH Headings
- Animals
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Mice
- Receptors, Neuropeptide/agonists
- Receptors, Neuropeptide/antagonists & inhibitors
- Receptors, Neuropeptide/metabolism
- Male
- Analgesics/pharmacology
- Analgesics/chemistry
- Analgesics/therapeutic use
- Analgesics/chemical synthesis
- Humans
- Structure-Activity Relationship
- Analgesics, Opioid/pharmacology
- Analgesics, Opioid/chemistry
Collapse
Affiliation(s)
- Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Khadija Elhabazi
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Simon Gonzalez
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Claire Herby
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Séverine Schneider
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Valérie Utard
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Rosine Fellmann-Clauss
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Nathalie Petit-Demouliere
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Sandra Lecat
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Mélanie Kremer
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (INCI), 67000 Strasbourg, France
| | - Aurelia Ces
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (INCI), 67000 Strasbourg, France
| | - François Daubeuf
- Plateforme de Chimie Biologique Intégrative de Strasbourg, UAR 3286, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Charlotte Martin
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Frédéric Bihel
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| |
Collapse
|
4
|
Blaine AT, van Rijn RM. Receptor expression and signaling properties in the brain, and structural ligand motifs that contribute to delta opioid receptor agonist-induced seizures. Neuropharmacology 2023; 232:109526. [PMID: 37004753 PMCID: PMC11078570 DOI: 10.1016/j.neuropharm.2023.109526] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 03/10/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
The δ opioid receptor (δOR) is a therapeutic target for the treatment of various neurological disorders, such as migraines, chronic pain, alcohol use, and mood disorders. Relative to μ opioid receptor agonists, δOR agonists show lower abuse liability and may be potentially safer analgesic alternatives. However, currently no δOR agonists are approved for clinical use. A small number of δOR agonists reached Phase II trials, but ultimately failed to progress due to lack of efficacy. One side effect of δOR agonism that remains poorly understood is the ability of δOR agonists to produce seizures. The lack of a clear mechanism of action is partly driven by the fact that δOR agonists range in their propensity to induce seizure behavior, with multiple δOR agonists reportedly not causing seizures. There is a significant gap in our current understanding of why certain δOR agonists are more likely to induce seizures, and what signal-transduction pathway and/or brain area is engaged to produce these seizures. In this review we provide a comprehensive overview of the current state of knowledge of δOR agonist-mediated seizures. The review was structured to highlight which agonists produce seizures, which brain regions have been implicated and which signaling mediators have been examined in this behavior. Our hope is that this review will spur future studies that are carefully designed and aimed to solve the question why certain δOR agonists are seizurogenic. Obtaining such insight may expedite the development of novel δOR clinical candidates without the risk of inducing seizures. This article is part of the Special Issue on "Opioid-induced changes in addiction and pain circuits".
Collapse
Affiliation(s)
- Arryn T Blaine
- Purdue University, Department of Medicinal Chemistry and Molecular Pharmacology, West Lafayette, IN, 47907, USA; Purdue University Interdisciplinary Life Science graduate program, West Lafayette, IN, 47907, USA
| | - Richard M van Rijn
- Purdue University, Department of Medicinal Chemistry and Molecular Pharmacology, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, West Lafayette, IN, 47907, USA; Purdue Institute for Drug Discovery, West Lafayette, IN, 47907, USA; Septerna Inc., South San Francisco, CA, 94080, USA.
| |
Collapse
|
5
|
Mei HR, Hu YY, Kapadia S, Ouimet T, Poras H, Dussor G. Efficacy of dual enkephalinase inhibition in a preclinical migraine model is mediated by activation of peripheral delta opioid receptors. Headache 2023; 63:621-633. [PMID: 37183526 PMCID: PMC10646790 DOI: 10.1111/head.14517] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/07/2023] [Accepted: 03/27/2023] [Indexed: 05/16/2023]
Abstract
OBJECTIVE The aim of this study was to evaluate whether elevating levels of enkephalin by inhibiting their degradation can attenuate stress-induced migraine-like behaviors in mice. BACKGROUND Previous studies in animals have suggested the delta opioid receptor (DOR) as a novel migraine target. The primary endogenous ligands for DOR are enkephalins and their levels can be increased by pharmacological inhibition of enkephalinases; however, it is not clear whether enkephalinase inhibition can be efficacious in preclinical migraine models through activation of DOR or whether other opioid receptors might be involved. Further, it is not clear whether opioid receptors in the central nervous system are necessary for these effects. METHODS This study used a model of repetitive restraint stress in mice that induces periorbital hypersensitivity and priming to the nitric oxide donor sodium nitroprusside (SNP; 0.1 mg/kg). Von Frey filaments were used to measure periorbital mechanical thresholds and grimace scores were evaluated by observing mouse facial features. Animals were treated with the dual enkephalinase inhibitor (DENKI) PL37. RESULTS On day two post-stress, PL37 given to mice via either intravenous injection (10 mg/kg) or oral gavage (20 mg/kg) significantly attenuated stress-induced periorbital hypersensitivity and facial grimace responses. Additionally, both intravenous (10 mg/kg) and oral gavage (20 mg/kg) of PL37 prior to SNP (0.1 mg/kg) administration on day 14 post-stress significantly reduced SNP-induced facial hypersensitivity. Injection of the DOR antagonist naltrindole (0.1 mg/kg) but not the mu-opioid receptor antagonist CTAP (1 mg/kg) prior to PL37 treatment blocked the effects. Finally, pretreatment of mice with the peripherally restricted opioid receptor antagonist naloxone methiodide (5 mg/kg) blocked the effects of PL37. CONCLUSIONS These data demonstrate that inhibiting enkephalinases, and thus protecting enkephalins from degradation, attenuates stress-induced migraine-like behavior via activation of peripheral DOR. Peripheral targeting of endogenous opioid signaling may be an effective therapeutic strategy for migraine.
Collapse
Affiliation(s)
- Hao-Ruei Mei
- Department of Neuroscience, University of Texas at Dallas, Texas, United States
- School of Behavioral and Brain Science, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States, United States
| | - Ya-Yu Hu
- Department of Neuroscience, University of Texas at Dallas, Texas, United States
- School of Behavioral and Brain Science, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States, United States
| | - Soneet Kapadia
- School of Behavioral and Brain Science, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States, United States
- Department of Biology and Biochemistry, University of Texas at Dallas, Texas, United States
| | | | | | - Gregory Dussor
- Department of Neuroscience, University of Texas at Dallas, Texas, United States
- School of Behavioral and Brain Science, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States, United States
| |
Collapse
|
6
|
Dremencov E, Grinchii D, Romanova Z, Chomanic P, Lacinova L, Jezova D. Effects of chronic delta-opioid receptor agonist on the excitability of hippocampal glutamate and brainstem monoamine neurons, anxiety, locomotion, and habituation in rats. Pharmacol Rep 2023; 75:585-595. [PMID: 37060527 DOI: 10.1007/s43440-023-00485-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND Short-term treatment with non-peptide agonists of delta-opioid receptors, such as agonist SNC80, induced behavioral effects in rodents, which could be modulated via changes in central neurotransmission. The present experiments aimed at testing the hypothesis that chronic treatment with SNC80 induces anxiolytic effects associated with changes in hippocampal glutamate and brainstem monoamine pathways. METHODS Adult male Wistar rats were used in experiments. Rats were treated with SNC80 (3 mg/kg/day) for fourteen days. Neuronal excitability was assessed using extracellular in vivo single-unit electrophysiology. The behavioral parameters were examined using the elevated plus maze and open field tests. RESULTS Chronic SNC80 treatment increased the excitability of hippocampal glutamate and ventral tegmental area dopamine neurons and had no effect on the firing activity of dorsal raphe nucleus serotonin cells. Chronic SNC80 treatment induced anxiolytic effects, which were, however, confounded by increased locomotor activity clearly confirmed in an open field test. The ability to cope with stressful situations and habituation processes in a novel environment was not influenced by chronic treatment with SNC80. CONCLUSION Our study suggests that the psychoactive effects of SNC80 might be explained by its ability to stimulate hippocampal glutamate and mesolimbic dopamine transmission.
Collapse
Affiliation(s)
- Eliyahu Dremencov
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia.
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Daniil Grinchii
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia
| | - Zuzana Romanova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
| | - Pavol Chomanic
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
| | - Lubica Lacinova
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
7
|
Olson KM, Hillhouse TM, Burgess GE, West JL, Hallahan JE, Dripps IJ, Ladetto AG, Rice KC, Jutkiewicz EM, Traynor JR. Delta Opioid Receptor-Mediated Antidepressant-Like Effects of Diprenorphine in Mice. J Pharmacol Exp Ther 2023; 384:343-352. [PMID: 36456196 PMCID: PMC9976798 DOI: 10.1124/jpet.122.001182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Major depressive disorder is a highly common disorder, with a lifetime prevalence in the United States of approximately 21%. Traditional antidepressant treatments are limited by a delayed onset of action and minimal efficacy in some patients. Ketamine is effective and fast-acting, but there are concerns over its abuse liability. Thus, there is a need for safe, fast-acting antidepressant drugs. The opioid buprenorphine shows promise but also has abuse liability due to its mu-agonist component. Preclinical evidence indicates that the delta-opioid system contributes to mood disorders, and delta-opioid agonists are effective in preclinical models of depression- and anxiety-like states. In this study, we test the hypothesis that the mu-opioid antagonist diprenorphine by virtue of its partial delta opioid agonist activity may offer a beneficial profile for an antidepressant medication without abuse liability. Diprenorphine was confirmed to bind with high affinity to all three opioid receptors, and functional experiments for G protein activation verified diprenorphine to be a partial agonist at delta- and kappa-opioid receptors and a mu-antagonist. Studies in C57BL/6 mice demonstrated that an acute dose of diprenorphine produced antidepressant-like effects in the tail suspension test and the novelty-induced hypophagia test that were inhibited in the presence of the delta-selective antagonist, naltrindole. Diprenorphine did not produce convulsions, a side effect of many delta agonists but rather inhibited convulsions caused by the full delta agonist SNC80; however, diprenorphine did potentiate pentylenetetrazole-induced convulsions. Diprenorphine, and compounds with a similar pharmacological profile, may provide efficient and safe rapidly acting antidepressants. SIGNIFICANCE STATEMENT: The management of major depressive disorder, particularly treatment-resistant depression, is a significant unmet medical need. Here we show that the opioid diprenorphine, a compound with mu-opioid receptor antagonist activity and delta- and kappa-opioid receptor partial agonist activities, has rapid onset antidepressant-like activity in animal models. Diprenorphine and compounds with a similar pharmacological profile to diprenorphine should be explored as novel antidepressant drugs.
Collapse
MESH Headings
- Animals
- Mice
- Analgesics, Opioid/pharmacology
- Antidepressive Agents/pharmacology
- Depressive Disorder, Major
- Diprenorphine/pharmacology
- Mice, Inbred C57BL
- Receptors, Opioid
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/metabolism
- Seizures/chemically induced
Collapse
Affiliation(s)
- Keith M Olson
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Todd M Hillhouse
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Gwendolyn E Burgess
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Joshua L West
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - James E Hallahan
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Isaac J Dripps
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Allison G Ladetto
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Kenner C Rice
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Emily M Jutkiewicz
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - John R Traynor
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| |
Collapse
|
8
|
Jin D, Chen H, Huang Y, Chen SR, Pan HL. δ-Opioid receptors in primary sensory neurons tonically restrain nociceptive input in chronic pain but do not enhance morphine analgesic tolerance. Neuropharmacology 2022; 217:109202. [PMID: 35917874 DOI: 10.1016/j.neuropharm.2022.109202] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/10/2022] [Accepted: 07/18/2022] [Indexed: 11/26/2022]
Abstract
δ-Opioid receptors (DORs, encoded by the Oprd1 gene) are expressed throughout the peripheral and central nervous system, and DOR stimulation reduces nociception. Previous studies suggest that DORs promote the development of analgesic tolerance of μ-opioid receptor (MOR) agonists. It is uncertain whether DORs expressed in primary sensory neurons are involved in regulating chronic pain and MOR agonist-induced tolerance. In this study, we generated Oprd1 conditional knockout (Oprd1-cKO) mice by crossing Advillin-Cre mice with Oprd1-floxed mice. DOR expression in the dorsal root ganglion was diminished in Oprd1-cKO mice. Systemic or intrathecal injection of the DOR agonist SNC-80 produced analgesia in wild-type (WT), but not Oprd1-cKO, mice. In contrast, intracerebroventricular injection of SNC-80 produced a similar analgesic effect in WT and Oprd1-cKO mice. However, morphine-induced analgesia, hyperalgesia, or analgesic tolerance did not differ between WT and Oprd1-cKO mice. Compared with WT mice, Oprd1-cKO mice showed increased mechanical and heat hypersensitivity after nerve injury or tissue inflammation. Furthermore, blocking DORs with naltrindole increased nociceptive sensitivity induced by nerve injury or tissue inflammation in WT, but not Oprd1-cKO, mice. In addition, naltrindole potentiated glutamatergic input from primary afferents to spinal dorsal horn neurons increased by nerve injury or CFA in WT mice; this effect was absent in Oprd1-cKO mice. Our findings indicate that DORs in primary sensory neurons are critically involved in the analgesic effect of DOR agonists but not morphine-induced analgesic tolerance. Presynaptic DORs at primary afferent central terminals constitutively inhibit inflammatory and neuropathic pain by restraining glutamatergic input to spinal dorsal horn neurons.
Collapse
Affiliation(s)
- Daozhong Jin
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hong Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yuying Huang
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
9
|
Low SA, Nielsen JJ, Coakley CM, Thomas M, Mbachu EU, Chen CL, Jones-Hall Y, Tremblay MI, Hicks JR, Low PS. An engineered dual function peptide to repair fractured bones. J Control Release 2022; 350:688-697. [PMID: 36030992 PMCID: PMC9897200 DOI: 10.1016/j.jconrel.2022.06.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 02/08/2023]
Abstract
Targeted drug delivery, often referred to as "smart" drug delivery, is a process whereby a therapeutic drug is delivered to specific parts of the body in a manner that increases its concentration at the desired sites relative to others. This approach is poised to revolutionize medicine as exemplified by the recent FDA approval of Cytalux (FDA approves pioneering drug for ovarian cancer surgery - Purdue University News) which is a folate-receptor targeted intraoperative near infrared (NIR) imaging agent that was developed in our laboratories. Fracture-associated morbidities and mortality affect a significant portion of world population. United states, Canada and Europe alone spent $48 billion in treating osteoporosis related fractures although this number doesn't count the economic burden due to loss in productivity. It is estimated that by 2050 ca 21 million hip fractures would occur globally which will be leading cause of premature death and disability. Despite the need for improvement in the treatment for fracture repair, methods for treating fractures have changed little in recent decades. Systemic delivery of fracture-homing bone anabolics holds great promise as a therapeutic strategy in this regard. Here we report the design of a fracture-targeted peptide comprised of a payload that binds and activates the parathyroid hormone receptor (PTHR1) and is linked to a targeting ligand comprised of 20 D-glutamic acids (D-Glu20) that directs accumulation of the payload specifically at fracture sites. This targeted delivery results in reduction of fracture healing times to <1/2 while creating repaired bones that are >2-fold stronger than saline-treated controls in mice. Moreover, this hydroxyapatite-targeted peptide can be administered without detectable toxicity to healthy tissues or modification of healthy bones in dogs. Additionally, since similar results are obtained upon treatment of osteoporotic and diabetic fractures in mice, and pain resolution is simultaneously accelerated by this approach, we conclude that this fracture-targeted anabolic peptide displays significant potential to revolutionize the treatment of bone fractures.
Collapse
Affiliation(s)
- Stewart A Low
- Department of Chemistry, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA.
| | - Jeffery J Nielsen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | | | - Mini Thomas
- Novosteo Inc., 1281 Win Hentschel Blvd, West Lafayette, IN 47906, USA
| | - Ephraim U Mbachu
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Christopher L Chen
- College of Health and Human Sciences, Purdue University, West Lafayette, IN, USA
| | - Yava Jones-Hall
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Madeleine I Tremblay
- Department of Chemistry, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Jonathan R Hicks
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA; Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
10
|
Blaine AT, Miao Y, Yuan J, Palant S, Liu RJ, Zhang ZY, van Rijn RM. Exploration of beta-arrestin isoform signaling pathways in delta opioid receptor agonist-induced convulsions. Front Pharmacol 2022; 13:914651. [PMID: 36059958 PMCID: PMC9428791 DOI: 10.3389/fphar.2022.914651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
The δ-opioid receptor (δOR) has been considered as a therapeutic target in multiple neurological and neuropsychiatric disorders particularly as δOR agonists are deemed safer alternatives relative to the more abuse-liable µ-opioid receptor drugs. Clinical development of δOR agonists, however, has been challenging in part due to the seizure-inducing effects of certain δOR agonists. Especially agonists that resemble the δOR-selective agonist SNC80 have well-established convulsive activity. Close inspection suggests that many of those seizurogenic δOR agonists efficaciously recruit β-arrestin, yet surprisingly, SNC80 displays enhanced seizure activity in β-arrestin 1 knockout mice. This finding led us to hypothesize that perhaps β-arrestin 1 is protective against, whereas β-arrestin 2 is detrimental for δOR-agonist-induced seizures. To investigate our hypothesis, we characterized three different δOR agonists (SNC80, ADL5859, ARM390) in cellular assays and in vivo in wild-type and β-arrestin 1 and β-arrestin 2 knockout mice for seizure activity. We also investigated downstream kinases associated with β-arrestin-dependent signal transduction. We discovered that δOR agonist-induced seizure activity strongly and positively correlates with β-arrestin 2 efficacy for the agonist, but that indirect inhibition of ERK activation using the MEK inhibitor SL327 did not inhibit seizure potency and duration. Inhibition of the PI3K/AKT/mTOR signaling with honokiol but not PQR530, attenuated SNC80 seizure duration in β-arrestin 1 knockout, but honokiol did not reduce SNC80-induced seizures in wild-type mice. Ultimately, our results indicate that β-arrestin 2 is correlated with δOR agonist-induced seizure intensity, but that global β-arrestin 1 knockout mice are a poor model system to investigate their mechanism of action.
Collapse
Affiliation(s)
- Arryn T. Blaine
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
- Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN, United States
| | - Yiming Miao
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Jinling Yuan
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Sophia Palant
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Rebecca J. Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
- Purdue Institute for Drug Discovery, West Lafayette, IN, United States
- Purdue University Cancer Center, West Lafayette, IN, United States
| | - Richard. M. van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
- Purdue Institute for Drug Discovery, West Lafayette, IN, United States
- Purdue University Cancer Center, West Lafayette, IN, United States
- Purdue Institute for Integrative Neuroscience, West Lafayette, IN, United States
- *Correspondence: Richard. M. van Rijn,
| |
Collapse
|
11
|
Raza Shah A, Rasool N, Bılal M, Mubarık A, Alı Hashmı M, Nadeem Akhtar M, Imran M, Ahmad G, Siddiqa A, Adnan Alı Shah S. Efficient Synthesis of 4‐Bromo‐
N
‐(1‐phenylethyl)benzamide, Arylation by Pd(0) Catalyst, Characterization and DFT Study. ChemistrySelect 2022. [DOI: 10.1002/slct.202200861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Alı Raza Shah
- Department of Chemistry Government College University Faisalabad 38000 Pakistan
| | - Nasır Rasool
- Department of Chemistry Government College University Faisalabad 38000 Pakistan
| | - Muhammad Bılal
- Department of Chemistry Government College University Faisalabad 38000 Pakistan
| | - Adeel Mubarık
- Department of Chemistry Government College University Faisalabad 38000 Pakistan
| | - Muhammad Alı Hashmı
- Department of Chemistry University of Education, Attock Campus Attock 43600 Pakistan
| | | | - Muhammad Imran
- Department of Chemistry, Faculty of Science King Khalid University P.O. Box 9004 Abha 61413 Saudi Arabia
- Research center for Advanced Materials Science (RCAMS) king Khalid University Abha 61514, P. O. Box 9004 Saudi Arabia
| | - Gulraız Ahmad
- Department of Chemistry Government College University Faisalabad 38000 Pakistan
| | - Ayesha Siddiqa
- Department of Chemistry Government College University Faisalabad 38000 Pakistan
| | - Syed Adnan Alı Shah
- Faculty of Pharmacy Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam Bandar Puncak Alam 42300 Malaysia
- Atta-ur-Rahman Institute for Natural Products Discovery (AuRIns) Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam Bandar Puncak Alam 42300 Malaysia
| |
Collapse
|
12
|
Bertels Z, Dripps IJ, Shah P, Moye LS, Tipton AF, Siegersma K, Pradhan AA. Delta opioid receptors in Nav1.8 expressing peripheral neurons partially regulate the effect of delta agonist in models of migraine and opioid-induced hyperalgesia. NEUROBIOLOGY OF PAIN 2022; 12:100099. [PMID: 35859654 PMCID: PMC9289726 DOI: 10.1016/j.ynpai.2022.100099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 11/07/2022]
Abstract
DOR in Nav1.8 cells do not regulate anti-migraine effects of DOR agonist. DOR in Nav1.8 cells is critical for effect of DOR agonist in peripheral OIH. DOR in Nav1.8 cells is not necessary for effect of DOR agonist in cephalic OIH.
Migraine is one of the most common pain disorders and causes disability in millions of people every year. Delta opioid receptors (DOR) have been identified as a novel therapeutic target for migraine and other headache disorders. DORs are present in both peripheral and central regions and it is unclear which receptor populations regulate migraine-associated effects. The aim of this study was to determine if DOR expressed in peripheral nociceptors regulates headache associated endpoints and the effect of delta agonists within these mouse models. We used a conditional knockout, in which DOR was selectively deleted from Nav1.8 expressing cells. Nav1.8-DOR mice and loxP control littermates were tested in models of chronic migraine-associated allodynia, opioid-induced hyperalgesia, migraine-associated negative affect, and aura. Nav1.8-DOR and loxP mice had comparable effect sizes in all of these models. The anti-allodynic effect of the DOR agonist, SNC80, was slightly diminished in the nitroglycerin model of migraine. Intriguingly, in the OIH model the peripheral effects of SNC80 were completely lost in Nav1.8-DOR mice while the cephalic effects remained intact. Regardless of genotype, SNC80 continued to inhibit conditioned place aversion associated with nitroglycerin and decreased cortical spreading depression events associated with migraine aura. These results suggest that DOR in Nav1.8-expressing nociceptors do not critically regulate the anti-migraine effects of delta agonist; and that brain-penetrant delta agonists would be a more effective drug development strategy.
Collapse
|
13
|
Chen Y, He Y, Zhao S, He X, Xue D, Xia Y. Hypoxic/Ischemic Inflammation, MicroRNAs and δ-Opioid Receptors: Hypoxia/Ischemia-Sensitive Versus-Insensitive Organs. Front Aging Neurosci 2022; 14:847374. [PMID: 35615595 PMCID: PMC9124822 DOI: 10.3389/fnagi.2022.847374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 03/21/2022] [Indexed: 11/15/2022] Open
Abstract
Hypoxia and ischemia cause inflammatory injury and critically participate in the pathogenesis of various diseases in various organs. However, the protective strategies against hypoxic and ischemic insults are very limited in clinical settings up to date. It is of utmost importance to improve our understanding of hypoxic/ischemic (H/I) inflammation and find novel therapies for better prevention/treatment of H/I injury. Recent studies provide strong evidence that the expression of microRNAs (miRNAs), which regulate gene expression and affect H/I inflammation through post-transcriptional mechanisms, are differentially altered in response to H/I stress, while δ-opioid receptors (DOR) play a protective role against H/I insults in different organs, including both H/I-sensitive organs (e.g., brain, kidney, and heart) and H/I-insensitive organs (e.g., liver and muscle). Indeed, many studies have demonstrated the crucial role of the DOR-mediated cyto-protection against H/I injury by several molecular pathways, including NLRP3 inflammasome modulated by miRNAs. In this review, we summarize our recent studies along with those of others worldwide, and compare the effects of DOR on H/I expression of miRNAs in H/I-sensitive and -insensitive organs. The alternation in miRNA expression profiles upon DOR activation and the potential impact on inflammatory injury in different organs under normoxic and hypoxic conditions are discussed at molecular and cellular levels. More in-depth investigations into this field may provide novel clues for new protective strategies against H/I inflammation in different types of organs.
Collapse
Affiliation(s)
- Yimeng Chen
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yichen He
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Shuchen Zhao
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaozhou He
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dong Xue
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- *Correspondence: Dong Xue,
| | - Ying Xia
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
- Ying Xia,
| |
Collapse
|
14
|
Wistrom E, Chase R, Smith PR, Campbell ZT. A compendium of validated pain genes. WIREs Mech Dis 2022; 14:e1570. [PMID: 35760453 PMCID: PMC9787016 DOI: 10.1002/wsbm.1570] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/28/2022] [Accepted: 06/06/2022] [Indexed: 12/30/2022]
Abstract
The development of novel pain therapeutics hinges on the identification and rigorous validation of potential targets. Model organisms provide a means to test the involvement of specific genes and regulatory elements in pain. Here we provide a list of genes linked to pain-associated behaviors. We capitalize on results spanning over three decades to identify a set of 242 genes. They support a remarkable diversity of functions spanning action potential propagation, immune response, GPCR signaling, enzymatic catalysis, nucleic acid regulation, and intercellular signaling. Making use of existing tissue and single-cell high-throughput RNA sequencing datasets, we examine their patterns of expression. For each gene class, we discuss archetypal members, with an emphasis on opportunities for additional experimentation. Finally, we discuss how powerful and increasingly ubiquitous forward genetic screening approaches could be used to improve our ability to identify pain genes. This article is categorized under: Neurological Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Eric Wistrom
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA
| | - Rebecca Chase
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA
| | - Patrick R. Smith
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA
| | - Zachary T. Campbell
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA,Center for Advanced Pain StudiesUniversity of Texas at DallasRichardsonTexasUSA
| |
Collapse
|
15
|
Paul AK, Smith CM, Rahmatullah M, Nissapatorn V, Wilairatana P, Spetea M, Gueven N, Dietis N. Opioid Analgesia and Opioid-Induced Adverse Effects: A Review. Pharmaceuticals (Basel) 2021; 14:1091. [PMID: 34832873 PMCID: PMC8620360 DOI: 10.3390/ph14111091] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 01/08/2023] Open
Abstract
Opioids are widely used as therapeutic agents against moderate to severe acute and chronic pain. Still, these classes of analgesic drugs have many potential limitations as they induce analgesic tolerance, addiction and numerous behavioural adverse effects that often result in patient non-compliance. As opium and opioids have been traditionally used as painkillers, the exact mechanisms of their adverse reactions over repeated use are multifactorial and not fully understood. Older adults suffer from cancer and non-cancer chronic pain more than younger adults, due to the physiological changes related to ageing and their reduced metabolic capabilities and thus show an increased number of adverse reactions to opioid drugs. All clinically used opioids are μ-opioid receptor agonists, and the major adverse effects are directly or potentially connected to this receptor. Multifunctional opioid ligands or peripherally restricted opioids may elicit fewer adverse effects, as shown in preclinical studies, but these results need reproducibility from further extensive clinical trials. The current review aims to overview various mechanisms involved in the adverse effects induced by opioids, to provide a better understanding of the underlying pathophysiology and, ultimately, to help develop an effective therapeutic strategy to better manage pain.
Collapse
Affiliation(s)
- Alok K. Paul
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7001, Australia;
| | - Craig M. Smith
- School of Medicine, Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC 3216, Australia;
| | - Mohammed Rahmatullah
- Department of Biotechnology & Genetic Engineering, University of Development Alternative, Dhanmondi, Dhaka 1207, Bangladesh;
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences, World Union for Herbal Drug Discovery (WUHeDD) and Research Excellence Center for Innovation and Health Products (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand;
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innrain 80–82, 6020 Innsbruck, Austria;
| | - Nuri Gueven
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7001, Australia;
| | - Nikolas Dietis
- Medical School, University of Cyprus, Nicosia 1678, Cyprus;
| |
Collapse
|
16
|
Reiss D, Maurin H, Audouard E, Martínez-Navarro M, Xue Y, Herault Y, Maldonado R, Cabañero D, Gaveriaux-Ruff C. Delta Opioid Receptor in Astrocytes Contributes to Neuropathic Cold Pain and Analgesic Tolerance in Female Mice. Front Cell Neurosci 2021; 15:745178. [PMID: 34602984 PMCID: PMC8483180 DOI: 10.3389/fncel.2021.745178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/23/2021] [Indexed: 01/13/2023] Open
Abstract
Background: The delta opioid receptor (DOR) contributes to pain control, and a major challenge is the identification of DOR populations that control pain, analgesia, and tolerance. Astrocytes are known as important cells in the pathophysiology of chronic pain, and many studies report an increased prevalence of pain in women. However, the implication of astrocytic DOR in neuropathic pain and analgesia, as well as the influence of sex in this receptor activity, remains unknown. Experimental Approach: We developed a novel conditional knockout (cKO) mouse line wherein DOR is deleted in astrocytes (named GFAP-DOR-KO), and investigated neuropathic mechanical allodynia as well as analgesia and analgesic tolerance in mutant male and female mice. Neuropathic cold allodynia was also characterized in mice of both sexes lacking DOR either in astrocytes or constitutively. Results: Neuropathic mechanical allodynia was similar in GFAP-DOR-KO and floxed DOR control mice, and the DOR agonist SNC80 produced analgesia in mutant mice of both sexes. Interestingly, analgesic tolerance developed in cKO males and was abolished in cKO females. Cold neuropathic allodynia was reduced in mice with decreased DOR in astrocytes. By contrast, cold allodynia was exacerbated in full DOR KO females. Conclusions: These findings show that astrocytic DOR has a prominent role in promoting cold allodynia and analgesic tolerance in females, while overall DOR activity was protective. Altogether this suggests that endogenous- and exogenous-mediated DOR activity in astrocytes worsens neuropathic allodynia while DOR activity in other cells attenuates this form of pain. In conclusion, our results show a sex-specific implication of astrocytic DOR in neuropathic pain and analgesic tolerance. These findings open new avenues for developing tailored DOR-mediated analgesic strategies.
Collapse
Affiliation(s)
- David Reiss
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Hervé Maurin
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Emilie Audouard
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Miriam Martínez-Navarro
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Yaping Xue
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Institute of Research, Development and Innovation in Healthcare Biotechnology of Elche (IDiBE), Universidad Miguel Hernández Elche, Alicante, Spain
| | - Claire Gaveriaux-Ruff
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| |
Collapse
|
17
|
Ko MJ, Chiang T, Mukadam AA, Mulia GE, Gutridge AM, Lin A, Chester JA, van Rijn RM. β-Arrestin-dependent ERK signaling reduces anxiety-like and conditioned fear-related behaviors in mice. Sci Signal 2021; 14:14/694/eaba0245. [PMID: 34344831 DOI: 10.1126/scisignal.aba0245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
G protein-coupled receptors (GPCRs) are implicated in the regulation of fear and anxiety. GPCR signaling involves canonical G protein pathways but can also engage downstream kinases and effectors through scaffolding interactions mediated by β-arrestin. Here, we investigated whether β-arrestin signaling regulates anxiety-like and fear-related behavior in mice in response to activation of the GPCR δ-opioid receptor (δOR or DOR). Administration of β-arrestin-biased δOR agonists to male C57BL/6 mice revealed β-arrestin 2-dependent activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in the dorsal hippocampus and amygdala and β-arrestin 1-dependent activation of ERK1/2 in the nucleus accumbens. In mice, β-arrestin-biased agonist treatment was associated with reduced anxiety-like and fear-related behaviors, with some overlapping and isoform-specific input. In contrast, applying a G protein-biased δOR agonist decreased ERK1/2 activity in all three regions as well as the dorsal striatum and was associated with increased fear-related behavior without effects on baseline anxiety. Our results indicate a complex picture of δOR neuromodulation in which β-arrestin 1- and 2-dependent ERK signaling in specific brain subregions suppresses behaviors associated with anxiety and fear and opposes the effects of G protein-biased signaling. Overall, our findings highlight the importance of noncanonical β-arrestin-dependent GPCR signaling in the regulation of these interrelated emotions.
Collapse
Affiliation(s)
- Mee Jung Ko
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA
| | - Terrance Chiang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Arbaaz A Mukadam
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Department of Psychological Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Grace E Mulia
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA
| | - Anna M Gutridge
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA
| | - Angel Lin
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Julia A Chester
- Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA.,Department of Psychological Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA. .,Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA.,Purdue Institute for Drug Discovery, West Lafayette, IN 47907, USA
| |
Collapse
|
18
|
Weiss N, Zamponi GW. Opioid Receptor Regulation of Neuronal Voltage-Gated Calcium Channels. Cell Mol Neurobiol 2021; 41:839-847. [PMID: 32514826 PMCID: PMC11448596 DOI: 10.1007/s10571-020-00894-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 12/28/2022]
Abstract
Neuronal voltage-gated calcium channels play a pivotal role in the conversion of electrical signals into calcium entry into nerve endings that is required for the release of neurotransmitters. They are under the control of a number of cellular signaling pathways that serve to fine tune synaptic activities, including G-protein coupled receptors (GPCRs) and the opioid system. Besides modulating channel activity via activation of second messengers, GPCRs also physically associate with calcium channels to regulate their function and expression at the plasma membrane. In this mini review, we discuss the mechanisms by which calcium channels are regulated by classical opioid and nociceptin receptors. We highlight the importance of this regulation in the control of neuronal functions and their implication in the development of disease conditions. Finally, we present recent literature concerning the use of novel μ-opioid receptor/nociceptin receptor modulators and discuss their use as potential drug candidates for the treatment of pain.
Collapse
Affiliation(s)
- Norbert Weiss
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
19
|
De Neve J, Barlow TMA, Tourwé D, Bihel F, Simonin F, Ballet S. Comprehensive overview of biased pharmacology at the opioid receptors: biased ligands and bias factors. RSC Med Chem 2021; 12:828-870. [PMID: 34223156 PMCID: PMC8221262 DOI: 10.1039/d1md00041a] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022] Open
Abstract
One of the main challenges in contemporary medicinal chemistry is the development of safer analgesics, used in the treatment of pain. Currently, moderate to severe pain is still treated with the "gold standard" opioids whose long-term often leads to severe side effects. With the discovery of biased agonism, the importance of this area of pharmacology has grown exponentially over the past decade. Of these side effects, tolerance, opioid misuse, physical dependence and substance use disorder (SUD) stand out, since these have led to many deaths over the past decades in both USA and Europe. New therapeutic molecules that induce a biased response at the opioid receptors (MOR, DOR, KOR and NOP receptor) are able to circumvent these side effects and, consequently, serve as more advantageous therapies with great promise. The concept of biased signaling extends far beyond the already sizeable field of GPCR pharmacology and covering everything would be vastly outside the scope of this review which consequently covers the biased ligands acting at the opioid family of receptors. The limitation of quantifying bias, however, makes this a controversial subject, where it is dependent on the reference ligand, the equation or the assay used for the quantification. Hence, the major issue in the field of biased ligands remains the translation of the in vitro profiles of biased signaling, with corresponding bias factors to in vivo profiles showing the presence or the lack of specific side effects. This review comprises a comprehensive overview of biased ligands in addition to their bias factors at individual members of the opioid family of receptors, as well as bifunctional ligands.
Collapse
Affiliation(s)
- Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Thomas M A Barlow
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Frédéric Bihel
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, CNRS Université de Strasbourg Illkirch France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242, CNRS, Université de Strasbourg Illkirch France
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| |
Collapse
|
20
|
Atigari DV, Paton KF, Uprety R, Váradi A, Alder AF, Scouller B, Miller JH, Majumdar S, Kivell BM. The mixed kappa and delta opioid receptor agonist, MP1104, attenuates chemotherapy-induced neuropathic pain. Neuropharmacology 2021; 185:108445. [PMID: 33383089 PMCID: PMC8344368 DOI: 10.1016/j.neuropharm.2020.108445] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/20/2020] [Accepted: 12/20/2020] [Indexed: 01/04/2023]
Abstract
Effective treatments for chronic pain without abuse liability are urgently needed. One in 5 adults suffer chronic pain and half of these patients report inefficient treatment. Mu opioid receptor agonists (MOP), including oxycodone, tramadol and morphine, are often prescribed to treat chronic pain, however, use of drugs targeting MOP can lead to drug dependency, tolerance and overdose deaths. Kappa opioid receptor (KOP) agonists have antinociceptive effects without abuse potential; however, they have not been utilised clinically due to dysphoria and sedation. We hypothesise that mixed opioid receptor agonists targeting the KOP and delta opioid receptor (DOP) would have a wider therapeutic index, with the rewarding effects of DOP negating the negative effects of KOP. MP1104, an analogue of 3-Iodobenzoyl naltrexamine, is a novel mixed opioid receptor agonist with potent antinociceptive effects mediated via KOP and DOP in mice without rewarding or aversive effects. In this study, we show MP1104 has potent, long-acting antinociceptive effects in the warm-water tail-withdrawal assay in male and female mice and rats; and is longer acting than morphine. In the paclitaxel-induced neuropathic pain model in mice, MP1104 reduced both mechanical and cold allodynia and unlike morphine, did not produce tolerance when administered daily for 23 days. Moreover, MP1104 did not induce sedative effects in the open-field locomotor activity test, respiratory depression in mice using whole-body plethysmography, or have cross-tolerance with morphine. This data supports the therapeutic development of mixed opioid receptor agonists, particularly mixed KOP/DOP agonists, as non-addictive pain medications with reduced tolerance.
Collapse
Affiliation(s)
- Diana Vivian Atigari
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Kelly Frances Paton
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Rajendra Uprety
- Molecular Pharmacology Program and Department of Neurology, Memorial Sloan Kettering Cancer Centre, New York, USA
| | - András Váradi
- Molecular Pharmacology Program and Department of Neurology, Memorial Sloan Kettering Cancer Centre, New York, USA
| | - Amy Frances Alder
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Brittany Scouller
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - John H Miller
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Susruta Majumdar
- Center of Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
| | - Bronwyn Maree Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand.
| |
Collapse
|
21
|
Dripps IJ, Bertels Z, Moye LS, Tipton AF, Siegersma K, Baca SM, Kieffer BL, Pradhan AA. Forebrain delta opioid receptors regulate the response of delta agonist in models of migraine and opioid-induced hyperalgesia. Sci Rep 2020; 10:17629. [PMID: 33077757 PMCID: PMC7573615 DOI: 10.1038/s41598-020-74605-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022] Open
Abstract
Delta opioid receptor (DOR) agonists have been identified as a promising novel therapy for headache disorders. DORs are broadly expressed in several peripheral and central regions important for pain processing and mood regulation; and it is unclear which receptors regulate headache associated symptoms. In a model of chronic migraine-associated pain using the human migraine trigger, nitroglycerin, we observed increased expression of DOR in cortex, hippocampus, and striatum; suggesting a role for these forebrain regions in the regulation of migraine. To test this hypothesis, we used conditional knockout mice with DORs deleted from forebrain GABAergic neurons (Dlx-DOR), and investigated the outcome of this knockout on the effectiveness of the DOR agonist SNC80 in multiple headache models. In DOR loxP controls SNC80 blocked the development of acute and chronic cephalic allodynia in the chronic nitroglycerin model, an effect that was lost in Dlx-DOR mice. In addition, the anti-allodynic effects of SNC80 were lost in a model of opioid induced hyperalgesia/medication overuse headache in Dlx-DOR conditional knockouts. In a model reflecting negative affect associated with migraine, SNC80 was only effective in loxP controls and not Dlx-DOR mice. Similarly, SNC80 was ineffective in the cortical spreading depression model of migraine aura in conditional knockout mice. Taken together, these data indicate that forebrain DORs are necessary for the action of DOR agonists in relieving headache-related symptoms and suggest that forebrain regions may play an important role in migraine modulation.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Analgesics, Opioid/therapeutic use
- Animals
- Benzamides/pharmacology
- Benzamides/therapeutic use
- Cortical Spreading Depression/drug effects
- Cortical Spreading Depression/physiology
- Disease Models, Animal
- GABAergic Neurons/drug effects
- GABAergic Neurons/metabolism
- Hyperalgesia/chemically induced
- Hyperalgesia/drug therapy
- Hyperalgesia/metabolism
- Mice
- Mice, Knockout
- Migraine Disorders/chemically induced
- Migraine Disorders/drug therapy
- Migraine Disorders/metabolism
- Nitroglycerin
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Prosencephalon/drug effects
- Prosencephalon/metabolism
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
Collapse
Affiliation(s)
- Isaac J Dripps
- Department of Psychiatry, University of Illinois at Chicago, 1601 W. Taylor Street (MC 912), Chicago, IL, 60612, USA
| | - Zachariah Bertels
- Department of Psychiatry, University of Illinois at Chicago, 1601 W. Taylor Street (MC 912), Chicago, IL, 60612, USA
| | - Laura S Moye
- Department of Psychiatry, University of Illinois at Chicago, 1601 W. Taylor Street (MC 912), Chicago, IL, 60612, USA
| | - Alycia F Tipton
- Department of Psychiatry, University of Illinois at Chicago, 1601 W. Taylor Street (MC 912), Chicago, IL, 60612, USA
| | - Kendra Siegersma
- Department of Psychiatry, University of Illinois at Chicago, 1601 W. Taylor Street (MC 912), Chicago, IL, 60612, USA
| | - Serapio M Baca
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, USA
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Brigitte L Kieffer
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Amynah A Pradhan
- Department of Psychiatry, University of Illinois at Chicago, 1601 W. Taylor Street (MC 912), Chicago, IL, 60612, USA.
| |
Collapse
|
22
|
Agonist-induced phosphorylation bar code and differential post-activation signaling of the delta opioid receptor revealed by phosphosite-specific antibodies. Sci Rep 2020; 10:8585. [PMID: 32444688 PMCID: PMC7244497 DOI: 10.1038/s41598-020-65589-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/05/2020] [Indexed: 01/08/2023] Open
Abstract
The δ-opioid receptor (DOP) is an attractive pharmacological target due to its potent analgesic, anxiolytic and anti-depressant activity in chronic pain models. However, some but not all selective DOP agonists also produce severe adverse effects such as seizures. Thus, the development of novel agonists requires a profound understanding of their effects on DOP phosphorylation, post-activation signaling and dephosphorylation. Here we show that agonist-induced DOP phosphorylation at threonine 361 (T361) and serine 363 (S363) proceeds with a temporal hierarchy, with S363 as primary site of phosphorylation. This phosphorylation is mediated by G protein-coupled receptor kinases 2 and 3 (GRK2/3) followed by DOP endocytosis and desensitization. DOP dephosphorylation occurs within minutes and is predominantly mediated by protein phosphatases (PP) 1α and 1β. A comparison of structurally diverse DOP agonists and clinically used opioids demonstrated high correlation between G protein-dependent signaling efficacies and receptor internalization. In vivo, DOP agonists induce receptor phosphorylation in a dose-dependent and agonist-selective manner that could be blocked by naltrexone in DOP-eGFP mice. Together, our studies provide novel tools and insights for ligand-activated DOP signaling in vitro and in vivo and suggest that DOP agonist efficacies may determine receptor post-activation signaling.
Collapse
|
23
|
Ceredig RA, Pierre F, Doridot S, Alduntzin U, Hener P, Salvat E, Yalcin I, Gaveriaux-Ruff C, Barrot M, Massotte D. Peripheral Delta Opioid Receptors Mediate Formoterol Anti-allodynic Effect in a Mouse Model of Neuropathic Pain. Front Mol Neurosci 2020; 12:324. [PMID: 32116538 PMCID: PMC7033630 DOI: 10.3389/fnmol.2019.00324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/17/2019] [Indexed: 01/18/2023] Open
Abstract
Neuropathic pain is a challenging condition for which current therapies often remain unsatisfactory. Chronic administration of β2 adrenergic agonists, including formoterol currently used to treat asthma and chronic obstructive pulmonary disease, alleviates mechanical allodynia in the sciatic nerve cuff model of neuropathic pain. The limited clinical data currently available also suggest that formoterol would be a suitable candidate for drug repurposing. The antiallodynic action of β2 adrenergic agonists is known to require activation of the delta-opioid (DOP) receptor but better knowledge of the molecular mechanisms involved is necessary. Using a mouse line in which DOP receptors were selectively ablated in neurons expressing Nav1.8 sodium channels (DOP cKO), we showed that these DOP peripheral receptors were necessary for the antiallodynic action of the β2 adrenergic agonist formoterol in the cuff model. Using a knock-in mouse line expressing a fluorescent version of the DOP receptor fused with the enhanced green fluorescent protein (DOPeGFP), we established in a previous study, that mechanical allodynia is associated with a smaller percentage of DOPeGFP positive small peptidergic sensory neurons in dorsal root ganglia (DRG), with a reduced density of DOPeGFP positive free nerve endings in the skin and with increased DOPeGFP expression at the cell surface. Here, we showed that the density of DOPeGFP positive free nerve endings in the skin is partially restored and no increase in DOPeGFP translocation to the plasma membrane is observed in mice in which mechanical pain is alleviated upon chronic oral administration of formoterol. This study, therefore, extends our previous results by confirming that changes in the mechanical threshold are associated with changes in peripheral DOP profile. It also highlights the common impact on DOP receptors between serotonin noradrenaline reuptake inhibitors such as duloxetine and the β2 mimetic formoterol.
Collapse
Affiliation(s)
- Rhian Alice Ceredig
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Florian Pierre
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Stéphane Doridot
- Chronobiotron, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Unai Alduntzin
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Pierre Hener
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Eric Salvat
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France.,Centre d'Evaluation et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Ipek Yalcin
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Claire Gaveriaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Université de Strasbourg, INSERM, Illkirch, France
| | - Michel Barrot
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Dominique Massotte
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
24
|
Abstract
Ischemic stroke is a global epidemic condition due to an inadequate supply of blood and oxygen to a specific area of brain either by arterial blockage or by narrowing of blood vessels. Despite having advancement in the use of thrombolytic and clot removal medicine, significant numbers of stroke patients are still left out without option for treatment. In this review, we summarize recent research work on the activation of δ-opioid receptor as a strategy for treating ischemic stroke-caused neuronal injury. Moreover, as activation of δ-opioid receptor by a non-peptidic δ-opioid receptor agonist also modulates the expression, maturation and processing of amyloid precursor protein and β-secretase activity, the potential role of these effects on ischemic stroke caused dementia or Alzheimer's disease are also discussed.
Collapse
Affiliation(s)
- Kalpana Subedi
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Hongmin Wang
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| |
Collapse
|
25
|
Nagase H, Saitoh A. Research and development of κ opioid receptor agonists and δ opioid receptor agonists. Pharmacol Ther 2019; 205:107427. [PMID: 31654658 DOI: 10.1016/j.pharmthera.2019.107427] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/30/2019] [Indexed: 12/28/2022]
Abstract
Delta opioid delta receptor (DOP) agonists were expected to be analgesics and many researchers tried to develop the SNC80 derivatives. However, the derivatives were dropped at the stage of early clinical trials because of undesirable side effects and weak analgesia. On the other hand, DOP agonists have been proposed as attractive candidates for the novel psychotropic drugs. We recently succeeded in synthesizing a novel selective DOP agonist KNT-127. KNT-127 produced neither catalepsy nor convulsive effects. We have demonstrated that KNT-127 has potent anxiolytic-like effect in rat models of innate anxiety. This anxiolytic-like effect was independent from known adverse effect of benzodiazepine, such as memory impairment, motor coordination deficits, and ethanol interactions. We have also demonstrated that KNT-127 showed potent and rapid antidepressant-like effects in rat models of depression. This antidepressant-like effect was independent from known adverse effect of selective serotonin reuptake inhibitor (SSRI), such as digestive symptoms. Therefore, we propose that DOP should be considered as an attractive target for the development of novel psychotropic drugs, without producing the adverse effects associated with benzodiazepine anxiolytics and SSRI antidepressants. Very recently, we developed another delta agonist NC-2800 with a different structure. NC-2800 is now in the preclinical stage using the CiCLE fund supported by AMED (Japanese Agency for Medical Research and Development).
Collapse
Affiliation(s)
- Hiroshi Nagase
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Akiyoshi Saitoh
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, 278-8510, Japan
| |
Collapse
|
26
|
Pan C, Meng H, Zhang S, Zuo Z, Shen Y, Wang L, Chang KJ. Homology modeling and 3D-QSAR study of benzhydrylpiperazine δ opioid receptor agonists. Comput Biol Chem 2019; 83:107109. [PMID: 31445419 DOI: 10.1016/j.compbiolchem.2019.107109] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/04/2019] [Accepted: 08/13/2019] [Indexed: 12/16/2022]
Abstract
The binding affinity of a series of benzhydrylpiperazine δ opioid receptor agonists were pooled and evaluated by using 3D-QSAR and homology modeling/molecular docking methods. Ligand-based CoMFA and CoMSIA 3D-QSAR analyses with 46 compounds were performed on benzhydrylpiperazine analogues by taking the most active compound BW373U86 as the template. The models were generated successfully with q2 value of 0.508 and r2 value of 0.964 for CoMFA, and q2 value of 0.530 and r2 value of 0.927 for CoMSIA. The predictive capabilities of the two models were validated on the test set with R2pred value of 0.720 and 0.814, respectively. The CoMSIA model appeared to work better in this case. A homology model of active form of δ opioid receptor was established by Swiss-Model using a reported crystal structure of active μ opioid receptor as a template, and was further optimized using nanosecond scale molecular dynamics simulation. The most active compound BW373U86 was docked to the active site of δ opioid receptor and the lowest energy binding pose was then used to identify binding residues such as s Gln105, Lys108, Leu125, Asp128, Tyr129, Leu200, Met132, Met199, Lys214, Trp274, Ile277, Ile304 and Tyr308. The docking and 3D-QSAR results showed that hydrogen bond and hydrophobic interactions played major roles in ligand-receptor interactions. Our results highlight that an approach combining structure-based homology modeling/molecular docking and ligand-based 3D-QSAR methods could be useful in designing of new opioid receptor agonists.
Collapse
Affiliation(s)
- Chenling Pan
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Hao Meng
- Beijing Beike Deyuan Bio-Pharm Technology Co., Ltd., Beijing, 100094, China
| | - Shuqun Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Zhili Zuo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Yuehai Shen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Liangliang Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Kwen-Jen Chang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
27
|
Receptor Ligands as Helping Hands to L-DOPA in the Treatment of Parkinson's Disease. Biomolecules 2019; 9:biom9040142. [PMID: 30970612 PMCID: PMC6523988 DOI: 10.3390/biom9040142] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/05/2019] [Accepted: 04/06/2019] [Indexed: 12/12/2022] Open
Abstract
Levodopa (LD) is the most effective drug in the treatment of Parkinson’s disease (PD). However, although it represents the “gold standard” of PD therapy, LD can cause side effects, including gastrointestinal and cardiovascular symptoms as well as transient elevated liver enzyme levels. Moreover, LD therapy leads to LD-induced dyskinesia (LID), a disabling motor complication that represents a major challenge for the clinical neurologist. Due to the many limitations associated with LD therapeutic use, other dopaminergic and non-dopaminergic drugs are being developed to optimize the treatment response. This review focuses on recent investigations about non-dopaminergic central nervous system (CNS) receptor ligands that have been identified to have therapeutic potential for the treatment of motor and non-motor symptoms of PD. In a different way, such agents may contribute to extending LD response and/or ameliorate LD-induced side effects.
Collapse
|
28
|
Usai EM, Manca I, Pettinau F, Mastino A, Pittau B. Chemical Characterization and in vitro
Metabolism of a Novel Class of Delta Opioid Receptor Agonists, Analogs of SNC-80. ChemistrySelect 2019. [DOI: 10.1002/slct.201803906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Elisabetta Maria Usai
- Institute of Translational Pharmacology; National Research Council; 09010 Pula (CA) Italy
| | - Ilaria Manca
- Institute of Translational Pharmacology; National Research Council; 09010 Pula (CA) Italy
| | - Francesca Pettinau
- Institute of Translational Pharmacology; National Research Council; 09010 Pula (CA) Italy
| | - Antonio Mastino
- Institute of Translational Pharmacology; National Research Council; 09010 Pula (CA) Italy
- Department of Chemical; Biological, Pharmaceutical, and Environmental Sciences; University of Messina; Messina Italy
| | - Barbara Pittau
- Institute of Translational Pharmacology; National Research Council; 09010 Pula (CA) Italy
| |
Collapse
|
29
|
Ceredig RA, Pierre F, Doridot S, Alduntzin U, Salvat E, Yalcin I, Gaveriaux-Ruff C, Barrot M, Massotte D. Peripheral delta opioid receptors mediate duloxetine antiallodynic effect in a mouse model of neuropathic pain. Eur J Neurosci 2018; 48:2231-2246. [PMID: 30059180 DOI: 10.1111/ejn.14093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/30/2018] [Accepted: 07/23/2018] [Indexed: 02/06/2023]
Abstract
Peripheral delta opioid (DOP) receptors are essential for the antiallodynic effect of the tricyclic antidepressant nortriptyline. However, the population of DOP-expressing cells affected in neuropathic conditions or underlying the antiallodynic activity of antidepressants remains unknown. Using a mouse line in which DOP receptors were selectively ablated in cells expressing Nav1.8 sodium channels (DOP cKO), we established that these DOP peripheral receptors were mandatory for duloxetine to alleviate mechanical allodynia in a neuropathic pain model based on sciatic nerve cuffing. We then examined the impact of nerve cuffing and duloxetine treatment on DOP-positive populations using a knock-in mouse line expressing a fluorescent version of the DOP receptor fused with the enhanced green fluorescent protein (DOPeGFP). Eight weeks postsurgery, we observed a reduced proportion of DOPeGFP-positive small peptidergic sensory neurons (calcitonin gene-related peptide (CGRP) positive) in dorsal root ganglia and a lower density of DOPeGFP-positive free nerve endings in the skin. These changes were not present in nerve-injured mice chronically treated with oral duloxetine. In addition, increased DOPeGFP translocation to the plasma membrane was observed in neuropathic conditions but not in duloxetine-treated neuropathic mice, which may represent an additional level of control of the neuronal activity by DOP receptors. Our results therefore established a parallel between changes in the expression profile of peripheral DOP receptors and mechanical allodynia induced by sciatic nerve cuffing.
Collapse
Affiliation(s)
- Rhian Alice Ceredig
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Florian Pierre
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Stéphane Doridot
- Centre National de la Recherche Scientifique, Chronobiotron, Strasbourg, France
| | - Unai Alduntzin
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Eric Salvat
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France.,Centre d'Evaluation et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Ipek Yalcin
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Claire Gaveriaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, Illkirch, France
| | - Michel Barrot
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Dominique Massotte
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
30
|
Sgroi S, Tonini R. Opioidergic Modulation of Striatal Circuits, Implications in Parkinson's Disease and Levodopa Induced Dyskinesia. Front Neurol 2018; 9:524. [PMID: 30026724 PMCID: PMC6041411 DOI: 10.3389/fneur.2018.00524] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/13/2018] [Indexed: 12/20/2022] Open
Abstract
The functional organization of the dorsal striatum is complex, due to the diversity of neural inputs that converge in this structure and its subdivision into direct and indirect output pathways, striosomes and matrix compartments. Among the neurotransmitters that regulate the activity of striatal projection neurons (SPNs), opioid neuropeptides (enkephalin and dynorphin) play a neuromodulatory role in synaptic transmission and plasticity and affect striatal-based behaviors in both normal brain function and pathological states, including Parkinson's disease (PD). We review recent findings on the cell-type-specific effects of opioidergic neurotransmission in the dorsal striatum, focusing on the maladaptive synaptic neuroadaptations that occur in PD and levodopa-induced dyskinesia. Understanding the plethora of molecular and synaptic mechanisms underpinning the opioid-mediated modulation of striatal circuits is critical for the development of pharmacological treatments that can alleviate motor dysfunctions and hyperkinetic responses to dopaminergic stimulant drugs.
Collapse
Affiliation(s)
- Stefania Sgroi
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Raffaella Tonini
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
31
|
Ogawa S, Natsume T, Takamatsu H. [Pharmacological profile of a novel nonhuman primate model of knee osteoarthritis]. Nihon Yakurigaku Zasshi 2018; 152:132-138. [PMID: 30185731 DOI: 10.1254/fpj.152.132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
A number of promising compounds developed in rodent arthritis models lack efficacy in clinical osteoarthritis (OA) pain. To enhance successful translation of preclinical findings, a nonhuman primate (NHP) model of knee OA was developed and characterized using behavioral assessments designed for use in the NHP. A unilateral medial meniscectomy (MMx) was performed and animals underwent an exercise regimen. Decreased ipsilateral knee pressure threshold, pressure "hyperalgesia", and decreased ipsilateral weight bearing, suggestive of pain at rest were observed. The sensitivity of the pain-related behaviors to pharmacological manipulation was evaluated. A single dose of the opioid morphine reduced pain-related behaviors. Likewise, the serotonin-norepinephrine reuptake inhibitor duloxetine reduced pain-related behavior, and efficacy was similar to that of morphine. By contrast, the anticonvulsant pregabalin did not significantly affect pain-related behavior. Repeated dosing with the non-steroidal anti-inflammatory drug (NSAID) diclofenac reduced pain-related behaviors whereas repeated dosing with the NK1 receptor antagonist aprepitant did not. The drug effects observed in the NHP OA model mirror the efficacy observed clinically.
Collapse
Affiliation(s)
- Shinya Ogawa
- Pharmacology Group, Hamamatsu Pharma Research, Inc
| | | | | |
Collapse
|
32
|
Lewis D. Biased for benefit: Stimulating the world's most popular drug targets with more nuance. Nat Med 2017; 23:649-651. [DOI: 10.1038/nm0617-649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
33
|
François A, Scherrer G. Delta Opioid Receptor Expression and Function in Primary Afferent Somatosensory Neurons. Handb Exp Pharmacol 2017; 247:87-114. [PMID: 28993838 DOI: 10.1007/164_2017_58] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The functional diversity of primary afferent neurons of the dorsal root ganglia (DRG) generates a variety of qualitatively and quantitatively distinct somatosensory experiences, from shooting pain to pleasant touch. In recent years, the identification of dozens of genetic markers specifically expressed by subpopulations of DRG neurons has dramatically improved our understanding of this diversity and provided the tools to manipulate their activity and uncover their molecular identity and function. Opioid receptors have long been known to be expressed by discrete populations of DRG neurons, in which they regulate cell excitability and neurotransmitter release. We review recent insights into the identity of the DRG neurons that express the delta opioid receptor (DOR) and the ion channel mechanisms that DOR engages in these cells to regulate sensory input. We highlight recent findings derived from DORGFP reporter mice and from in situ hybridization and RNA sequencing studies in wild-type mice that revealed DOR presence in cutaneous mechanosensory afferents eliciting touch and implicated in tactile allodynia. Mechanistically, we describe how DOR modulates opening of voltage-gated calcium channels (VGCCs) to control glutamatergic neurotransmission between somatosensory neurons and postsynaptic neurons in the spinal cord dorsal horn. We additionally discuss other potential signaling mechanisms, including those involving potassium channels, which DOR may engage to fine tune somatosensation. We conclude by discussing how this knowledge may explain the analgesic properties of DOR agonists against mechanical pain and uncovers an unanticipated specialized function for DOR in cutaneous mechanosensation.
Collapse
Affiliation(s)
- Amaury François
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.,Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.,Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA. .,Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA. .,Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
34
|
Chiang T, Sansuk K, van Rijn RM. β-Arrestin 2 dependence of δ opioid receptor agonists is correlated with alcohol intake. Br J Pharmacol 2016; 173:332-43. [PMID: 26507558 DOI: 10.1111/bph.13374] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/30/2015] [Accepted: 10/11/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE δ Opioid receptor agonists are being developed as potential treatments for depression and alcohol use disorders. This is particularly interesting as depression is frequently co-morbid with alcohol use disorders. Yet we have previously shown that δ receptor agonists range widely in their ability to modulate alcohol intake; certain δ receptor agonists actually increase alcohol consumption in mice. We propose that variations in β-arrestin 2 recruitment contribute to the differential behavioural profile of δ receptor agonists. EXPERIMENTAL APPROACH We used three diarylmethylpiperazine-based non-peptidic δ receptor selective agonists (SNC80, SNC162 and ARM390) and three structurally diverse δ receptor agonists (TAN-67, KNT127 and NIH11082). We tested these agonists in cAMP and β-arrestin 2 recruitment assays and a behavioural assay of alcohol intake in male C57BL/6 mice. We used β-arrestin 2 knockout mice and a model of depression-like behaviour to further study the role of β-arrestin 2 in δ receptor pharmacology. KEY RESULTS All six tested δ receptor agonists were full agonists in the cAMP assay but displayed distinct β-arrestin 2 recruitment efficacy. The efficacy of δ receptor agonists to recruit β-arrestin 2 positively correlated with their ability to increase alcohol intake (P < 0.01). The effects of the very efficacious recruiter SNC80 on alcohol intake, alcohol place preference and depression-like behaviour were β-arrestin 2-dependent. CONCLUSIONS AND IMPLICATIONS Our finding that δ receptor agonists that strongly recruit β-arrestin 2 can increase alcohol intake carries important ramifications for drug development of δ receptor agonists for treatment of alcohol use disorders and depressive disorders.
Collapse
Affiliation(s)
- T Chiang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | | | | |
Collapse
|
35
|
Vicario N, Parenti R, Arico' G, Turnaturi R, Scoto GM, Chiechio S, Parenti C. Repeated activation of delta opiod receptors counteracts nerve injury-induced TNF-α up-regulation in the sciatic nerve of rats with neuropathic pain: A possible correlation with delta opiod receptors-mediated antiallodinic effect. Mol Pain 2016; 12:12/0/1744806916667949. [PMID: 27590071 PMCID: PMC5024981 DOI: 10.1177/1744806916667949] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Despite mu opioid receptor agonists are the cornerstones of moderate-to-severe acute pain treatment, their effectiveness in chronic pain conditions is controversial. In contrast to mu opioid receptor agonists, a number of studies have reported the effectiveness of delta opioid receptor agonists on neuropathic pain strengthening the idea that delta opioid receptors gain importance when chronic pain develops. Among other effects, it has been shown that delta opioid receptor activation in optic nerve astrocytes inhibits tumor necrosis factor-α-mediated inflammation in response to severe hypoxia. Considering the involvement of tumor necrosis factor-α in the development and maintenance of neuropathic pain, with this study we sought to correlate the effect of delta opioid receptor agonist on the development of mechanical allodynia to tumor necrosis factor-α expression at the site of nerve injury in rats subjected to chronic constriction injury of the sciatic nerve. To this aim, we measured the levels of tumor necrosis factor-α in the sciatic nerve of rats with neuropathic pain after repeated injections with a delta opioid receptor agonist. Results obtained demonstrated that repeated administrations of the delta opioid receptor agonist SNC80 (10 mg/kg, i.p. for seven consecutive days) significantly inhibited the development of mechanical allodynia in rats with neuropathic pain and that the improvement of neuropathic symptom was timely related to the reduced expression of tumor necrosis factor-α in the rat sciatic nerve. We demonstrated also that when treatment with the delta opioid receptor agonist was suspended both allodynia and tumor necrosis factor-α up-regulation in the sciatic nerve of rats with neuropathic pain were restored. These results show that persistent delta opioid receptor activation significantly attenuates neuropathic pain and negatively regulates sciatic nerve tumor necrosis factor-α expression in chronic constriction injury rats.
Collapse
Affiliation(s)
| | | | | | | | | | - Santina Chiechio
- University of CataniaUniversity of CataniaUniversity of CataniaUniversity of CataniaUniversity of Catania
| | - Carmela Parenti
- University of CataniaUniversity of CataniaUniversity of CataniaUniversity of CataniaUniversity of Catania
| |
Collapse
|
36
|
Gendron L, Cahill CM, von Zastrow M, Schiller PW, Pineyro G. Molecular Pharmacology of δ-Opioid Receptors. Pharmacol Rev 2016; 68:631-700. [PMID: 27343248 PMCID: PMC4931872 DOI: 10.1124/pr.114.008979] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Opioids are among the most effective analgesics available and are the first choice in the treatment of acute severe pain. However, partial efficacy, a tendency to produce tolerance, and a host of ill-tolerated side effects make clinically available opioids less effective in the management of chronic pain syndromes. Given that most therapeutic opioids produce their actions via µ-opioid receptors (MOPrs), other targets are constantly being explored, among which δ-opioid receptors (DOPrs) are being increasingly considered as promising alternatives. This review addresses DOPrs from the perspective of cellular and molecular determinants of their pharmacological diversity. Thus, DOPr ligands are examined in terms of structural and functional variety, DOPrs' capacity to engage a multiplicity of canonical and noncanonical G protein-dependent responses is surveyed, and evidence supporting ligand-specific signaling and regulation is analyzed. Pharmacological DOPr subtypes are examined in light of the ability of DOPr to organize into multimeric arrays and to adopt multiple active conformations as well as differences in ligand kinetics. Current knowledge on DOPr targeting to the membrane is examined as a means of understanding how these receptors are especially active in chronic pain management. Insight into cellular and molecular mechanisms of pharmacological diversity should guide the rational design of more effective, longer-lasting, and better-tolerated opioid analgesics for chronic pain management.
Collapse
Affiliation(s)
- Louis Gendron
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Catherine M Cahill
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Mark von Zastrow
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Peter W Schiller
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Graciela Pineyro
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| |
Collapse
|
37
|
Paeoniflorin exerts analgesic and hypnotic effects via adenosine A1 receptors in a mouse neuropathic pain model. Psychopharmacology (Berl) 2016; 233:281-93. [PMID: 26514553 DOI: 10.1007/s00213-015-4108-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/05/2015] [Indexed: 10/22/2022]
Abstract
RATIONAL Neuropathic pain is frequently comorbid with sleep disturbances. Paeoniflorin, a main active compound of total glucosides of paeony, has been well documented to exhibit neuroprotective bioactivity. OBJECTIVE The present study evaluated effects of paeoniflorin on neuropathic pain and associated insomnia and the mechanisms involved. METHODS The analgesic and hypnotic effects of paeoniflorin were measured by mechanical threshold and thermal latency, electroencephalogram (EEG) and electromyogram, and c-Fos expression in a neuropathic pain insomnia model. RESULTS The data revealed that paeoniflorin (50 or 100 mg/kg, i.p.) significantly increased the mechanical threshold and prolonged the thermal latency in partial sciatic nerve ligation (PSNL) mice. Meanwhile, paeoniflorin increased non-rapid eye movement (NREM) sleep amount and concomitantly decreased wakefulness time. However, pretreatment with l,3-dimethy-8-cyclopenthylxanthine, an adenosine A1 receptor (R, A1R) antagonist, abolished the analgesic and hypnotic effects of paeoniflorin. Moreover, paeoniflorin at 100 mg/kg failed to change mechanical threshold and thermal latency and NREM sleep in A1R knockout PSNL mice. Immunohistochemical study showed that paeoniflorin inhibited c-Fos overexpression induced by PSNL in the anterior cingulate cortex and ventrolateral periaqueductal gray. CONCLUSIONS The present findings indicated that paeoniflorin exerted analgesic and hypnotic effects via adenosine A1Rs and might be of potential use in the treatment of neuropathic pain and associated insomnia.
Collapse
|
38
|
Pellissier LP, Pujol CN, Becker JAJ, Le Merrer J. Delta Opioid Receptors: Learning and Motivation. Handb Exp Pharmacol 2016; 247:227-260. [PMID: 28035528 DOI: 10.1007/164_2016_89] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Delta opioid receptor (DOR) displays a unique, highly conserved, structure and an original pattern of distribution in the central nervous system, pointing to a distinct and specific functional role among opioid peptide receptors. Over the last 15 years, in vivo pharmacology and genetic models have allowed significant advances in the understanding of this role. In this review, we will focus on the involvement of DOR in modulating different types of hippocampal- and striatal-dependent learning processes as well as motor function, motivation, and reward. Remarkably, DOR seems to play a key role in balancing hippocampal and striatal functions, with major implications for the control of cognitive performance and motor function under healthy and pathological conditions.
Collapse
Affiliation(s)
- L P Pellissier
- Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, INSERM, Université François Rabelais, IFCE, 37380, Nouzilly, France
| | - C N Pujol
- Département de Neurosciences, Institut de Génomique fonctionnelle, INSERM U-661, CNRS UMR-5203, 34094, Montpellier, France
| | - J A J Becker
- Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, INSERM, Université François Rabelais, IFCE, 37380, Nouzilly, France
| | - J Le Merrer
- Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, INSERM, Université François Rabelais, IFCE, 37380, Nouzilly, France.
| |
Collapse
|
39
|
Saitoh A, Nagase H. Delta Opioid Receptor (DOR) Ligands and Pharmacology: Development of Indolo- and Quinolinomorphinan Derivatives Based on the Message-Address Concept. Handb Exp Pharmacol 2016; 247:3-19. [PMID: 27787711 DOI: 10.1007/164_2016_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The pharmacology of the delta opioid receptor (DOR) has lagged, mainly due to the lack of an agonist with high potency and selectivity in vivo. The DOR is now receiving increasing attention, and there has been progress in the synthesis of better novel ligands. The discovery of a selective receptor DOR antagonist, naltrindole (NTI), stimulated the design and synthesis of (±)TAN-67, which was designed based on the message-address concept and the accessory site theory. Intensive studies using (±)TAN-67 determined the DOR-mediated various pharmacological effects, such as antinociceptive effects for painful diabetic neuropathy and cardiovascular protective effects. We improved the agonist activity of TAN-67 to afford SN-28, which was modified to KNT-127, a novel compound that improved the blood-brain barrier permeability. In addition, KNT-127 showed higher selectivity for the DOR and had potent agonist activity following systemic administration. Interestingly, KNT-127 produced no convulsive effects, unlike prototype DOR agonists. The KNT-127 type derivatives with a quinolinomorphinan structure are expected to be promising candidates for the development of therapeutic DOR agonists.
Collapse
Affiliation(s)
- Akiyoshi Saitoh
- Department of Neuropsychopharmacology, National Center of Neurology and Psychiatry, National Institute of Mental Health, Tokyo, 187-8553, Japan
| | - Hiroshi Nagase
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
40
|
Peppin JF, Raffa RB. Delta opioid agonists: a concise update on potential therapeutic applications. J Clin Pharm Ther 2015; 40:155-66. [PMID: 25726896 DOI: 10.1111/jcpt.12244] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/24/2014] [Indexed: 01/23/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE The endogenous opioid system co-evolved with chemical defences, or at times symbiotic relationships, between plants and other autotrophs and heterotrophic predators - thus, it is not surprising that endogenous opioid ligands and exogenous mimetic ligands produce diverse physiological effects. Among the endogenous opioid peptides (endomorphins, enkephalins, dynorphins and nociception/orphanin FQ) derived from the precursors encoded by four genes (PNOC, PENK, PDYN and POMC) are the pentapeptides Met-enkephalin (Tyr-Gly-Gly-Phe-Met) and Leu-enkephalin (Tyr-Gly-Gly-Phe-Leu). The physiological effects of the enkephalins are mediated via 7-transmembrane G protein-coupled receptors, including delta opioid receptor (DOR). We present a concise update on the status of progress and opportunities of this approach. METHODS A literature search of the PUBMED database and a combination of keywords including delta opioid receptor, analgesia, mood and individual compounds identified therein, from industry and other source, and from www.clinicaltrials.com. RESULTS AND DISCUSSION DOR agonist and antagonist ligands have been developed with ever increasing affinity and selectivity for DOR over other opioid receptor subtypes and studied for therapeutic utility, primarily for pain relief, but also for other clinical endpoints. WHAT IS NEW AND CONCLUSION Selective DOR agonists have been designed with a large increase in therapeutic window for a variety of potential CNS applications including pain, depression, and learning and memory among others.
Collapse
Affiliation(s)
- J F Peppin
- Center for Bioethics, Pain Management and Medicine, University City, MO, USA; Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | | |
Collapse
|
41
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 790] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|
42
|
Wang K, Xu J, Hunter DJ, Ding C. Investigational drugs for the treatment of osteoarthritis. Expert Opin Investig Drugs 2015; 24:1539-1556. [PMID: 26429673 DOI: 10.1517/13543784.2015.1091880] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Osteoarthritis (OA) is a common joint disease with multiple pathophysiological processes, affecting the whole joint. Current therapeutic options such as NSAIDs can provide a palliative effect on symptoms but have limited effect on disease progression. New drugs targeting OA structures may retard disease progression at an earlier stage and delay the need for joint replacement. AREAS COVERED Some drugs have entered into clinical trials and a few, such as strontium ranelate, do have improvements in both pain and structure changes. However, most of them have failed in clinical trials largely due to increased side effects or the failure to identify the right OA phenotype for the right drug in clinical design. This review describes various investigational drugs developed for the treatment of OA covering those at stages from preclinical experiments to early phase clinical trials. They include drugs for slowing cartilage degradation, regulating cartilage metabolism, targeting subchondral bone, controlling inflammation and relieving pain. EXPERT OPINION Treatment options for OA remain limited. However, with the emergence of sensitive tools to detect early disease progression and identification of different OA phenotypes, disease-modifying anti-OA drugs with increased benefit and reduced risks will become available for OA treatment in the near future.
Collapse
Affiliation(s)
- Kang Wang
- a 1 The First Affiliated Hospital of Anhui Medical University, Arthritis Research Institute, Department of Rheumatology and Immunology , Hefei, China
| | - Jianhua Xu
- a 1 The First Affiliated Hospital of Anhui Medical University, Arthritis Research Institute, Department of Rheumatology and Immunology , Hefei, China
| | - David J Hunter
- b 2 University of Sydney, Kolling Institute, Institute of Bone and Joint Research, Royal North Shore Hospital, Rheumatology Department , Sydney, NSW, Australia
| | - Changhai Ding
- a 1 The First Affiliated Hospital of Anhui Medical University, Arthritis Research Institute, Department of Rheumatology and Immunology , Hefei, China
- c 3 University of Tasmania, Menzies Institute for Medical Research , Private Bag 23, Hobart, Tasmania 7000, Australia +61 3 62 26 77 30 ; +61 3 62 26 77 04 ;
| |
Collapse
|
43
|
Loriga G, Lazzari P, Manca I, Ruiu S, Falzoi M, Murineddu G, Bottazzi MEH, Pinna G, Pinna GA. Novel diazabicycloalkane delta opioid agonists. Bioorg Med Chem 2015; 23:5527-38. [DOI: 10.1016/j.bmc.2015.07.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 07/17/2015] [Accepted: 07/19/2015] [Indexed: 12/19/2022]
|
44
|
Nozaki C, Nagase H, Nemoto T, Matifas A, Kieffer BL, Gaveriaux-Ruff C. In vivo properties of KNT-127, a novel δ opioid receptor agonist: receptor internalization, antihyperalgesia and antidepressant effects in mice. Br J Pharmacol 2015; 171:5376-86. [PMID: 25048778 DOI: 10.1111/bph.12852] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 06/30/2014] [Accepted: 07/13/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Activation of δ opioid (DOP) receptors regulates pain and emotional responses, and also displays ligand-biased agonism. KNT-127 (1,2,3,4,4a,5,12,12a-octahydro-2-methyl-4aβ,1β-([1,2]benzenomethano)-2,6-diazanaphthacene-12aβ,17-diol) is a novel DOP receptor agonist inducing analgesia and antidepressant effects in mice. Here, we have assessed KNT-127 for (i) analgesia against chronic inflammatory pain; (ii) effects on depression, locomotion and DOP receptor internalization; and (iii) for cross-tolerance to analgesic and antidepressant effects of acute treatment by other DOP receptor agonists. EXPERIMENTAL APPROACH Inflammatory pain was induced by complete Freund's adjuvant injection into tail or hindpaw, and thermal and mechanical sensitivities were determined in mice. Locomotor and antidepressant-like effects were measured using actimetry and forced swim test respectively. In vivo KNT-127 selectivity and internalization were assessed using DOP receptor knockout mice and knock-in mice expressing fluorescent-tagged DOP receptors. KNT-127 was injected acutely at 0.1-10.0 mg·kg(-1) or administered chronically at 5 mg·kg(-1) daily over 5 days. KEY RESULTS Acute treatment with KNT-127 reversed inflammatory hyperalgesia, produced an antidepressant-like effect but induced neither hyperlocomotion nor receptor sequestration. Chronic treatment with KNT-127 induced tolerance and cross-tolerance to SNC80-induced analgesia, but no tolerance to SNC80-evoked hyperlocomotor or antidepressant-like effects. CONCLUSIONS AND IMPLICATIONS The DOP receptor agonist KNT-127 induced agonist-specific acute and chronic responses, at both behavioural and cellular levels. It displays activities similar to the other recently reported DOP agonists, AR-M1000390, ADL5747 and ADL5859, and differs from SNC80. SNC80 differs from the other DOP receptor agonists including KNT-127, by exhibiting ligand-biased tolerance at this receptor.
Collapse
Affiliation(s)
- C Nozaki
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany; Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Strasbourg, France
| | | | | | | | | | | |
Collapse
|
45
|
Gibula-Bruzda E, Marszalek-Grabska M, Witkowska E, Izdebski J, Kotlinska JH. Enkephalin analog, cyclo[N(ε),N(β)-carbonyl-D-Lys(2),Dap(5)] enkephalinamide (cUENK6), inhibits the ethanol withdrawal-induced anxiety-like behavior in rats. Alcohol 2015; 49:229-36. [PMID: 25716198 DOI: 10.1016/j.alcohol.2015.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 01/20/2015] [Accepted: 01/22/2015] [Indexed: 01/08/2023]
Abstract
An analog of enkephalin, cyclo[N(ε),N(β)-carbonyl-D-Lys(2),Dap(5)] enkephalinamide (cUENK6), is predominantly a functional agonist of μ-opioid receptors (MOPr) and, to a lesser extent, of δ-opioid receptors (DOPr) in vitro. The aim of the present study was to determine whether cUENK6 could affect ethanol withdrawal-induced anxiety-like behavior in the elevated plus maze (EPM) test in rats. An anxiety-like effect of withdrawal was predicted to occur in the EPM test 24 h after the last ethanol administration (2 g/kg, intraperitoneally [i.p.]; 15% w/v once daily for 9 days). Ethanol withdrawal decreased the percent of time spent by rats in the open arms and the percent of open-arms entries. cUENK6 (0.25 nmol), given by intracerebroventricular (i.c.v.) injection, significantly reversed these anxiety-like effects of ethanol withdrawal and elevated the percent of time spent by rats in the open arms and the percent of open-arms entries. These effects of cUENK6 were significantly inhibited by the DOPr antagonist naltrindole (NTI) (5 nmol, i.c.v.), but not by the MOPr antagonist β-funaltrexamine (β-FNA) (5 nmol, i.c.v.). The preferential DOPr agonist [Leu(5)]-enkephalin (LeuEnk) (2.7 and 5.4 nmol, i.c.v.) and the MOPr agonist morphine (6.5 and 13 nmol, i.c.v.) reduced the anxiety-like effects of ethanol withdrawal. cUENK6 at the dose of 0.25 nmol did not disturb locomotor activity in the EPM, in contrast to cUENK6 at the dose of 0.5 nmol, and morphine at 6.5 and 13 nmol. However, similarly to LeuEnk, cUENK6 induced the anxiolytic-like effects in naïve rats. Thus, our study suggests that cUENK6 reduced ethanol withdrawal-induced anxiety-like behavior by activation of δ-opioid receptors rather than μ-opioid receptors.
Collapse
|
46
|
A novel anxiogenic role for the delta opioid receptor expressed in GABAergic forebrain neurons. Biol Psychiatry 2015; 77:404-15. [PMID: 25444168 PMCID: PMC4297504 DOI: 10.1016/j.biopsych.2014.07.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 07/15/2014] [Accepted: 07/24/2014] [Indexed: 01/10/2023]
Abstract
BACKGROUND The delta opioid receptor (DOR) is broadly expressed throughout the nervous system; it regulates chronic pain, emotional responses, motivation, and memory. Neural circuits underlying DOR activities have been poorly explored by genetic approaches. We used conditional mouse mutagenesis to elucidate receptor function in GABAergic neurons of the forebrain. METHODS We characterized DOR distribution in the brain of Dlx5/6-CreXOprd1(fl/fl) (Dlx-DOR) mice and tested main central DOR functions through behavioral testing. RESULTS The DOR proteins were strongly deleted in olfactory bulb and striatum and remained intact in cortex and basolateral amygdala. Olfactory perception, circadian activity, and despair-like behaviors were unchanged. In contrast, locomotor stimulant effects of SNC80 (DOR agonist) and SKF81297 (D1 agonist) were abolished and increased, respectively. The Dlx-DOR mice showed lower levels of anxiety in the elevated plus maze, opposing the known high anxiety in constitutive DOR knockout animals. Also, Dlx-DOR mice reached the food more rapidly in a novelty suppressed feeding task, despite their lower motivation for food reward observed in an operant paradigm. Finally, c-fos protein staining after novelty suppressed feeding was strongly reduced in amygdala, concordant with the low anxiety phenotype of Dlx-DOR mice. CONCLUSIONS We demonstrate that DORs expressed in the forebrain mediate the described locomotor effect of SNC80 and inhibit D1-stimulated hyperactivity. Our data also reveal an unanticipated anxiogenic role for this particular DOR subpopulation, with a potential novel adaptive role. In emotional responses, DORs exert dual anxiolytic and anxiogenic roles, both of which may have implications in the area of anxiety disorders.
Collapse
|
47
|
Chung PCS, Boehrer A, Stephan A, Matifas A, Scherrer G, Darcq E, Befort K, Kieffer BL. Delta opioid receptors expressed in forebrain GABAergic neurons are responsible for SNC80-induced seizures. Behav Brain Res 2015; 278:429-34. [PMID: 25447299 PMCID: PMC4382405 DOI: 10.1016/j.bbr.2014.10.029] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 10/12/2014] [Accepted: 10/21/2014] [Indexed: 01/01/2023]
Abstract
The delta opioid receptor (DOR) has raised much interest for the development of new therapeutic drugs, particularly to treat patients suffering from mood disorders and chronic pain. Unfortunately, the prototypal DOR agonist SNC80 induces mild epileptic seizures in rodents. Although recently developed agonists do not seem to show convulsant properties, mechanisms and neuronal circuits that support DOR-mediated epileptic seizures remain to be clarified. DORs are expressed throughout the nervous system. In this study we tested the hypothesis that SNC80-evoked seizures stem from DOR activity at the level of forebrain GABAergic transmission, whose inhibition is known to facilitate the development of epileptic seizures. We generated a conditional DOR knockout mouse line, targeting the receptor gene specifically in GABAergic neurons of the forebrain (Dlx-DOR). We measured effects of SNC80 (4.5, 9, 13.5 and 32 mg/kg), ARM390 (10, 30 and 60 mg/kg) or ADL5859 (30, 100 and 300 mg/kg) administration on electroencephalograms (EEGs) recorded in Dlx-DOR mice and their control littermates (Ctrl mice). SNC80 produced dose-dependent seizure events in Ctrl mice, but these effects were not detected in Dlx-DOR mice. As expected, ARM390 and ADL5859 did not trigger any detectable change in mice from both genotypes. These results demonstrate for the first time that SNC80-induced DOR activation induces epileptic seizures via direct inhibition of GABAergic forebrain neurons, and supports the notion of differential activities between first and second-generation DOR agonists.
Collapse
Affiliation(s)
- Paul Chu Sin Chung
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Annie Boehrer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Aline Stephan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Audrey Matifas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Molecular and Cellular Physiology, Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Emmanuel Darcq
- Douglas Hospital Research Center, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Katia Befort
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Brigitte L Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France; Douglas Hospital Research Center, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
48
|
Megat S, Bohren Y, Doridot S, Gaveriaux-Ruff C, Kieffer BL, Freund-Mercier MJ, Yalcin I, Barrot M. κ-Opioid receptors are not necessary for the antidepressant treatment of neuropathic pain. Br J Pharmacol 2014; 172:1034-44. [PMID: 25297905 DOI: 10.1111/bph.12963] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 09/22/2014] [Accepted: 09/26/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Tricyclic antidepressants are used clinically as first-line treatments for neuropathic pain. Opioid receptors participate in this pain-relieving action, and preclinical studies in receptor-deficient mice have highlighted a critical role for δ-, but not μ-opioid receptors. In this study, we investigated whether κ-opioid (KOP) receptors have a role in the antiallodynic action of tricyclic antidepressants. EXPERIMENTAL APPROACH We used a model of neuropathic pain induced by unilateral sciatic nerve cuffing. In this model, the mechanical allodynia was evaluated using von Frey filaments. Experiments were conducted in C57BL/6J mice, and in KOP receptor-deficient mice and their wild-type littermates. The tricyclic antidepressant nortriptyline (5 mg · kg(-1)) was delivered twice a day for over 2 weeks. Agonists and antagonists of opioid receptors were used to test the selectivity of the KOP receptor antagonist norbinaltorphimine (nor-BNI) in mice with neuropathic pain. KEY RESULTS After 12 days of treatment, nortriptyline relieved neuropathic allodynia in both wild-type and KOP receptor-deficient mice. Surprisingly, acute nor-BNI reversed the effect of nortriptyline in both wild-type and KOP receptor-deficient mice. Further experiments showed that nor-BNI action was selective for KOP receptors at a late time-point after its administration (8 h), but not at an early time-point, when it may also interact with δ-opioid (DOP) receptors. CONCLUSIONS AND IMPLICATIONS KOP receptors are not necessary for the effect of a tricyclic antidepressant against neuropathic allodynia. These findings together with previous data indicate that the DOP receptor is the only opioid receptor that is necessary for the antiallodynic action of antidepressants.
Collapse
Affiliation(s)
- Salim Megat
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France; Université de Strasbourg, Strasbourg, France
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Adeghate E, Fehér E, Kalász H. Evaluating the Phase II drugs currently under investigation for diabetic neuropathy. Expert Opin Investig Drugs 2014; 24:1-15. [PMID: 25171371 DOI: 10.1517/13543784.2014.954033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Introduction: The worldwide number of patients suffering from diabetes mellitus (DM) is projected to approach 552 million by the year 2030. As diabetic neuropathy (DN) is present in 8% of new diabetic patients at the time of diagnosis and occurs in ∼ 50% of all patients with established DM, the number of patients who will develop painful DN will also increase. The suboptimal efficacies of currently approved drugs have prompted investigators to develop new therapeutic agents for the management of painful DN. Areas covered: In this review, the authors present and elucidate the current status of drugs under investigation for the treatment of painful DN. A short synopsis of currently approved drugs is also given. Literature information and data analysis were retrieved from PubMed, the American Diabetes and Neurological Associations Websites and ClinicalTrials.gov. The keywords used in the search included: DM, DN, painful diabetic neuropathy. Expert opinion: In addition to treating the pain associated with DN, the actual causes of the disease should also be targeted for improved management. It is hoped that drugs which improve vascular blood flow, induce neural regeneration, reduce hyperglycemia, oxidative stress and inflammation can be more effective for the overall treatment of painful DN.
Collapse
Affiliation(s)
- Ernest Adeghate
- United Arab Emirates University, College of Medicine and Health Sciences, Department of Anatomy , P.O Box 17666, Al Ain , UAE +971 3 7672033 ;
| | | | | |
Collapse
|
50
|
Mika J, Popiolek-Barczyk K, Rojewska E, Makuch W, Starowicz K, Przewlocka B. Delta-opioid receptor analgesia is independent of microglial activation in a rat model of neuropathic pain. PLoS One 2014; 9:e104420. [PMID: 25105291 PMCID: PMC4126741 DOI: 10.1371/journal.pone.0104420] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/09/2014] [Indexed: 12/15/2022] Open
Abstract
The analgesic effect of delta-opioid receptor (DOR) ligands in neuropathic pain is not diminished in contrast to other opioid receptor ligands, which lose their effectiveness as analgesics. In this study, we examine whether this effect is related to nerve injury-induced microglial activation. We therefore investigated the influence of minocycline-induced inhibition of microglial activation on the analgesic effects of opioid receptor agonists: morphine, DAMGO, U50,488H, DPDPE, Deltorphin II and SNC80 after chronic constriction injury (CCI) to the sciatic nerve in rats. Pre-emptive and repeated administration of minocycline (30 mg/kg, i.p.) over 7 days significantly reduced allodynia and hyperalgesia as measured on day 7 after CCI. The antiallodynic and antihyperalgesic effects of intrathecally (i.t.) administered morphine (10–20 µg), DAMGO (1–2 µg) and U50,488H (25–50 µg) were significantly potentiated in rats after minocycline, but no such changes were observed after DPDPE (10–20 µg), deltorphin II (1.5–15 µg) and SNC80 (10–20 µg) administration. Additionally, nerve injury-induced down-regulation of all types of opioid receptors in the spinal cord and dorsal root ganglia was not influenced by minocycline, which indicates that the effects of opioid ligands are dependent on other changes, presumably neuroimmune interactions. Our study of rat primary microglial cell culture using qRT-PCR, Western blotting and immunocytochemistry confirmed the presence of mu-opioid receptors (MOR) and kappa-opioid receptors (KOR), further we provide the first evidence for the lack of DOR on microglial cells. In summary, DOR analgesia is different from analgesia induced by MOR and KOR receptors because it does not dependent on injury-induced microglial activation. DOR agonists appear to be the best candidates for new drugs to treat neuropathic pain.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/therapeutic use
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/therapeutic use
- Animals
- Anti-Bacterial Agents/administration & dosage
- Anti-Bacterial Agents/therapeutic use
- Cells, Cultured
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/therapeutic use
- Gene Expression Regulation/drug effects
- Male
- Microglia/cytology
- Microglia/drug effects
- Microglia/metabolism
- Minocycline/administration & dosage
- Minocycline/therapeutic use
- Morphine/administration & dosage
- Morphine/therapeutic use
- Neuralgia/drug therapy
- Rats, Wistar
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
Collapse
Affiliation(s)
- Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
- * E-mail: (BP); (JM)
| | | | - Ewelina Rojewska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wioletta Makuch
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Starowicz
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Barbara Przewlocka
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
- * E-mail: (BP); (JM)
| |
Collapse
|