1
|
Drakopoulos A, Koszegi Z, Seier K, Hübner H, Maurel D, Sounier R, Granier S, Gmeiner P, Calebiro D, Decker M. Design, Synthesis, and Characterization of New δ Opioid Receptor-Selective Fluorescent Probes and Applications in Single-Molecule Microscopy of Wild-Type Receptors. J Med Chem 2024; 67:12618-12631. [PMID: 39044606 PMCID: PMC11386433 DOI: 10.1021/acs.jmedchem.4c00627] [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] [Indexed: 07/25/2024]
Abstract
The delta opioid receptor (δOR or DOR) is a G protein-coupled receptor (GPCR) showing a promising profile as a drug target for nociception and analgesia. Herein, we design and synthesize new fluorescent antagonist probes with high δOR selectivity that are ideally suited for single-molecule microscopy (SMM) applications in unmodified, untagged receptors. Using our new probes, we investigated wild-type δOR localization and mobility at low physiological receptor densities for the first time. Furthermore, we investigate the potential formation of δOR homodimers, as such a receptor organization might exhibit distinct pharmacological activity, potentially paving the way for innovative pharmacological therapies. Our findings indicate that the majority of δORs labeled with these probes exist as freely diffusing monomers on the cell surface in a simple cell model. This discovery advances our understanding of OR behavior and offers potential implications for future therapeutic research.
Collapse
Affiliation(s)
- Antonios Drakopoulos
- Pharmazeutische und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Julius-Maximilians-Universität (JMU) Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Zsombor Koszegi
- Institute of Metabolism and Systems Research, University of Birmingham, B15 2TT Birmingham, U.K
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, B15 2TT Birmingham, U.K
| | - Kerstin Seier
- Institute of Pharmacology and Toxicology, Julius-Maximilians University of Würzburg, Versbacher Strasse 9, 97078 Würzburg, Germany
| | - Harald Hübner
- Chair of Pharmaceutical Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander University of Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Damien Maurel
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, 34094 Cedex 5 Montpellier, France
| | - Rémy Sounier
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, 34094 Cedex 5 Montpellier, France
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, 34094 Cedex 5 Montpellier, France
| | - Peter Gmeiner
- Chair of Pharmaceutical Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander University of Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Davide Calebiro
- Institute of Metabolism and Systems Research, University of Birmingham, B15 2TT Birmingham, U.K
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, B15 2TT Birmingham, U.K
| | - Michael Decker
- Pharmazeutische und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Julius-Maximilians-Universität (JMU) Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
2
|
Ganeyan A, Ganesh CB. Organization of enkephalinergic neuronal system in the central nervous system of the gecko Hemidactylus frenatus. Brain Struct Funct 2024; 229:1365-1395. [PMID: 38713249 DOI: 10.1007/s00429-024-02805-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
Enkephalins are endogenous opioid pentapeptides that play a role in neurotransmission and pain modulation in vertebrates. However, the distribution pattern of enkephalinergic neurons in the brains of reptiles has been understudied. This study reports the organization of the methionine-enkephalin (M-ENK) and leucine-enkephalin (L-ENK) neuronal systems in the central nervous system of the gecko Hemidactylus frenatus using an immunofluorescence labeling method. Although M-ENK and L-ENK-immunoreactive (ir) fibers extended throughout the pallial and subpallial subdivisions, including the olfactory bulbs, M-ENK and L-ENK-ir cells were found only in the dorsal septal nucleus. Enkephalinergic perikarya and fibers were highly concentrated in the periventricular and lateral preoptic areas, as well as in the anterior and lateral subdivisions of the hypothalamus, while enkephalinergic innervation was observed in the hypothalamic periventricular nucleus, infundibular recess nucleus and median eminence. The dense accumulation of enkephalinergic content was noticed in the pars distalis of the hypophysis. In the thalamus, the nucleus rotundus and the dorsolateral, medial, and medial posterior thalamic nuclei contained M-ENK and L-ENK-ir fibers, whereas clusters of M-ENK and L-ENK-ir neurons were observed in the pretectum, mesencephalon, and rhombencephalon. The enkephalinergic fibers were also seen in the area X around the central canal, as well as the dorsal and ventral horns. The widespread distribution of enkephalin-containing neurons within the central nervous system implies that enkephalins regulate a variety of functions in the gecko, including sensory, behavioral, hypophysiotropic, and neuroendocrine functions.
Collapse
Affiliation(s)
- Ananya Ganeyan
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad, 580 003, India
| | - C B Ganesh
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad, 580 003, India.
| |
Collapse
|
3
|
Meqbil YJ, Aguilar J, Blaine AT, Chen L, Cassell RJ, Pradhan AA, van Rijn RM. Identification of 1,3,8-Triazaspiro[4.5]Decane-2,4-Dione Derivatives as a Novel δ Opioid Receptor-Selective Agonist Chemotype. J Pharmacol Exp Ther 2024; 389:301-309. [PMID: 38621994 PMCID: PMC11125782 DOI: 10.1124/jpet.123.001735] [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: 05/10/2023] [Revised: 03/25/2024] [Accepted: 04/08/2024] [Indexed: 04/17/2024] Open
Abstract
δ opioid receptors (DORs) hold potential as a target for neurologic and psychiatric disorders, yet no DOR agonist has proven efficacious in critical phase II clinical trials. The exact reasons for the failure to produce quality drug candidates for the DOR are unclear. However, it is known that certain DOR agonists can induce seizures and exhibit tachyphylaxis. Several studies have suggested that those adverse effects are more prevalent in delta agonists that share the (+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide (SNC80)/4-[(αR*)-α-((2S*,5R*)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-hydroxybenzyl]-N,N-diethylbenzamide chemotype. There is a need to find novel lead candidates for drug development that have improved pharmacological properties to differentiate them from the current failed delta agonists. Our objective in this study was to identify novel DOR agonists. We used a β-arrestin assay to screen a small G-protein coupled receptors (GPCR)-focused chemical library. We identified a novel chemotype of DOR agonists that appears to bind to the orthosteric site based of docking and molecular dynamic simulation. The most potent agonist hit compound is selective for the DOR over a panel of 167 other GPCRs, is slightly biased toward G-protein signaling and has anti-allodynic efficacy in a complete Freund's adjuvant model of inflammatory pain in C57BL/6 male and female mice. The newly discovered chemotype contrasts with molecules like SNC80 that are highly efficacious β-arrestin recruiters and may suggest this novel class of DOR agonists could be expanded on to develop a clinical candidate drug. SIGNIFICANCE STATEMENT: δ opioid receptors are a clinical target for various neurological disorders, including migraine and chronic pain. Many of the clinically tested delta opioid agonists share a single chemotype, which carries risks during drug development. Through a small-scale high-throughput screening assay, this study identified a novel δ opioid receptor agonist chemotype, which may serve as alternative for the current analgesic clinical candidates.
Collapse
Affiliation(s)
- Yazan J Meqbil
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Jhoan Aguilar
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Arryn T Blaine
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Lan Chen
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Robert J Cassell
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Amynah A Pradhan
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Richard M van Rijn
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| |
Collapse
|
4
|
Silvestro M, Iannone LF, Orologio I, Tessitore A, Tedeschi G, Geppetti P, Russo A. Migraine Treatment: Towards New Pharmacological Targets. Int J Mol Sci 2023; 24:12268. [PMID: 37569648 PMCID: PMC10418850 DOI: 10.3390/ijms241512268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Migraine is a debilitating neurological condition affecting millions of people worldwide. Until a few years ago, preventive migraine treatments were based on molecules with pleiotropic targets, developed for other indications, and discovered by serendipity to be effective in migraine prevention, although often burdened by tolerability issues leading to low adherence. However, the progresses in unravelling the migraine pathophysiology allowed identifying novel putative targets as calcitonin gene-related peptide (CGRP). Nevertheless, despite the revolution brought by CGRP monoclonal antibodies and gepants, a significant percentage of patients still remains burdened by an unsatisfactory response, suggesting that other pathways may play a critical role, with an extent of involvement varying among different migraine patients. Specifically, neuropeptides of the CGRP family, such as adrenomedullin and amylin; molecules of the secretin family, such as pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP); receptors, such as transient receptor potential (TRP) channels; intracellular downstream determinants, such as potassium channels, but also the opioid system and the purinergic pathway, have been suggested to be involved in migraine pathophysiology. The present review provides an overview of these pathways, highlighting, based on preclinical and clinical evidence, as well as provocative studies, their potential role as future targets for migraine preventive treatment.
Collapse
Affiliation(s)
- Marcello Silvestro
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (I.O.); (A.T.); (G.T.)
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Luigi Francesco Iannone
- Headache Centre and Clinical Pharmacology Unit, Careggi University Hospital Florence, 50134 Florence, Italy; (L.F.I.); (P.G.)
| | - Ilaria Orologio
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (I.O.); (A.T.); (G.T.)
| | - Alessandro Tessitore
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (I.O.); (A.T.); (G.T.)
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Gioacchino Tedeschi
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (I.O.); (A.T.); (G.T.)
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Pierangelo Geppetti
- Headache Centre and Clinical Pharmacology Unit, Careggi University Hospital Florence, 50134 Florence, Italy; (L.F.I.); (P.G.)
| | - Antonio Russo
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| |
Collapse
|
5
|
Cahill CM, Holdridge SV, Liu S, Xue L, Magnussen C, Ong E, Grenier P, Sutherland A, Olmstead MC. Delta opioid receptor activation modulates affective pain and modality-specific pain hypersensitivity associated with chronic neuropathic pain. J Neurosci Res 2022; 100:129-148. [PMID: 32623788 PMCID: PMC8218601 DOI: 10.1002/jnr.24680] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/28/2020] [Accepted: 06/04/2020] [Indexed: 01/03/2023]
Abstract
Delta opioid receptor (DOR) agonists alleviate nociceptive behaviors in various chronic pain models, including neuropathic pain, while having minimal effect on sensory thresholds in the absence of injury. The mechanisms underlying nerve injury-induced enhancement of DOR function are unclear. We used a peripheral nerve injury (PNI) model of neuropathic pain to assess changes in the function and localization of DORs in mice and rats. Intrathecal administration of DOR agonists reversed mechanical allodynia and thermal hyperalgesia. The dose-dependent thermal antinociceptive effects of DOR agonists were shifted to the left in PNI rats. Administration of DOR agonists produced a conditioned place preference in PNI, but not in sham, animals, whereas the DOR antagonist naltrindole produced a place aversion in PNI, but not in sham, mice, suggesting the engagement of endogenous DOR activity in suppressing pain associated with the injury. GTPγS autoradiography revealed an increase in DOR function in the dorsal spinal cord, ipsilateral to PNI. Immunogold electron microscopy and in vivo fluorescent agonist assays were used to assess changes in the ultrastructural localization of DORs in the spinal dorsal horn. In shams, DORs were primarily localized within intracellular compartments. PNI significantly increased the cell surface expression of DORs within lamina IV-V dendritic profiles. Using neonatal capsaicin treatment, we identified that DOR agonist-induced thermal antinociception was mediated via receptors expressed on primary afferent sensory neurons but did not alter mechanical thresholds. These data reveal that the regulation of DORs following PNI and suggest the importance of endogenous activation of DORs in regulating chronic pain states.
Collapse
Affiliation(s)
- Catherine M. Cahill
- Dept of Psychiatry & Biobehavioral Sciences, Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, USA, 90095
| | - Sarah V. Holdridge
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Steve Liu
- Dept of Psychiatry & Biobehavioral Sciences, Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, USA, 90095,Department of Psychology and Centre for Neuroscience Studies, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Lihua Xue
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Claire Magnussen
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Edmund Ong
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Patrick Grenier
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Anne Sutherland
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Mary C. Olmstead
- Department of Psychology and Centre for Neuroscience Studies, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| |
Collapse
|
6
|
Xu Y, Chen R, Zhi F, Sheng S, Khiati L, Yang Y, Peng Y, Xia Y. δ-opioid Receptor, Microglia and Neuroinflammation. Aging Dis 2022; 14:778-793. [PMID: 37191426 DOI: 10.14336/ad.2022.0912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/12/2022] [Indexed: 11/18/2022] Open
Abstract
Neuroinflammation underlies the pathophysiology of multiple age-related neurological disorders. Microglia, the resident immune cells of the central nervous system, are critically involved in neuroinflammatory regulation and neural survival. Modulating microglial activation is thus a promising approach to alleviate neuronal injury. Our serial studies have revealed a neuroprotective role of the δ-opioid receptor (DOR) in several acute and chronic cerebral injuries by regulating neuroinflammation and cellular oxidative stress. More recently, we found an endogenous mechanism for the inhibition of neuroinflammation is closely related to DOR's modulation of microglia. Our recent studies showed that DOR activation could strongly protect neurons from hypoxia- and lipopolysaccharide (LPS)-induced injury by inhibiting microglial pro-inflammatory transformation, while knocking-down DOR or restraining DOR activity promoted microglia activation and the relevant inflammatory events with an aggravation of cell injury. This novel finding highlights a therapeutic potential of DOR in numerous age-related neurological disorders through the modulation of neuroinflammation by targeting microglia. This review summarized the current data regarding the role of microglia in neuroinflammation, oxidative stress, and age-related neurological diseases focusing on the pharmacological effects and signaling transduction of DOR in microglia.
Collapse
|
7
|
Piras S, Murineddu G, Loriga G, Carta A, Battistello E, Merighi S, Gessi S, Corona P, Asproni B, Ibba R, Temml V, Schuster D, Pinna GA. Biological Effects on μ-Receptors Affinity and Selectivity of Arylpropenyl Chain Structural Modification on Diazatricyclodecane Derivatives. Molecules 2021; 26:5448. [PMID: 34576918 PMCID: PMC8467848 DOI: 10.3390/molecules26185448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Opioid analgesics are clinically used to relieve severe pain in acute postoperative and cancer pain, and also in the long term in chronic pain. The analgesic action is mediated by μ-, δ-, and κ-receptors, but currently, with few exceptions for k-agonists, μ-agonists are the only ones used in therapy. Previously synthesized compounds with diazotricyclodecane cores (DTDs) have shown their effectiveness in binding opioid receptors. Fourteen novel diazatricyclodecanes belonging to the 9-propionyl-10-substituted-9,10-diazatricyclo[4.2.1.12,5]decane (compounds 20-23, 53, 57 and 59) and 2-propionyl-7-substituted-2,7-diazatricyclo[4.4.0.03,8]decane (compounds 24-27, 54, 58 and 60) series, respectively, have been synthesized and their ability to bind to the opioid μ-, δ- and κ-receptors was evaluated. Five of these derivatives, compounds 20, 21, 24, 26 and 53, showed μ-affinity in the nanomolar range with a negligible affinity towards δ- and κ-receptors and high μ-receptor selectivity. The synthesized compounds showed μ-receptor selectivity higher than those of previously reported methylarylcinnamyl analogs.
Collapse
Affiliation(s)
- Sandra Piras
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Gabriele Murineddu
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Giovanni Loriga
- Institute of Biomolecular Chemistry, National Research Council, Traversa La Crucca 3, 07100 Sassari, Italy;
| | - Antonio Carta
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Enrica Battistello
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.B.); (S.M.); (S.G.)
| | - Stefania Merighi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.B.); (S.M.); (S.G.)
| | - Stefania Gessi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.B.); (S.M.); (S.G.)
| | - Paola Corona
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Battistina Asproni
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Roberta Ibba
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Veronika Temml
- Department of Pharmaceutical Chemistry, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; (V.T.); (D.S.)
| | - Daniela Schuster
- Department of Pharmaceutical Chemistry, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; (V.T.); (D.S.)
| | - Gérard Aimè Pinna
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| |
Collapse
|
8
|
Moye LS, Siegersma K, Dripps I, Witkowski W, Mangutov E, Wang D, Scherrer G, Pradhan AA. Delta opioid receptor regulation of calcitonin gene-related peptide dynamics in the trigeminal complex. Pain 2021; 162:2297-2308. [PMID: 33605657 PMCID: PMC8730473 DOI: 10.1097/j.pain.0000000000002235] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022]
Abstract
ABSTRACT Migraine is highly prevalent and is the sixth leading cause worldwide for years lost to disability. Therapeutic options specifically targeting migraine are limited, and delta opioid receptor (DOP) agonists were recently identified as a promising pharmacotherapy. The mechanisms by which DOPs regulate migraine are currently unclear. Calcitonin gene-related peptide (CGRP) has been identified as an endogenous migraine trigger and plays a critical role in migraine initiation and susceptibility. The aim of this study was to determine the behavioral effects of DOP agonists on the development of chronic migraine-associated pain and to investigate DOP coexpression with CGRP and CGRP receptor (CGRPR) in the trigeminal system. Chronic migraine-associated pain was induced in mice through repeated intermittent injection of the known human migraine trigger, nitroglycerin. Chronic nitroglycerin resulted in severe chronic cephalic allodynia which was prevented with cotreatment of the DOP-selective agonist, SNC80. In addition, a corresponding increase in CGRP expression in the trigeminal ganglia and trigeminal nucleus caudalis was observed after chronic nitroglycerin, an augmentation that was blocked by SNC80. Moreover, DOP was also upregulated in these head pain-processing regions following the chronic migraine model. Immunohistochemical analysis of the trigeminal ganglia revealed coexpression of DOP with CGRP as well as with a primary component of the CGRPR, RAMP1. In the trigeminal nucleus caudalis, DOP was not coexpressed with CGRP but was highly coexpressed with RAMP1 and calcitonin receptor-like receptor. These results suggest that DOP agonists inhibit migraine-associated pain by attenuating CGRP release and blocking pronociceptive signaling of the CGRPR.
Collapse
Affiliation(s)
- Laura S Moye
- Department of Psychiatry, University of Illinois at Chicago
| | | | - Isaac Dripps
- Department of Psychiatry, University of Illinois at Chicago
| | | | | | - Dong Wang
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Neurosurgery, Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- New York Stem Cell Foundation – Robertson Investigator
| | | |
Collapse
|
9
|
Pasquinucci L, Parenti C, Georgoussi Z, Reina L, Tomarchio E, Turnaturi R. LP1 and LP2: Dual-Target MOPr/DOPr Ligands as Drug Candidates for Persistent Pain Relief. Molecules 2021; 26:molecules26144168. [PMID: 34299443 PMCID: PMC8305117 DOI: 10.3390/molecules26144168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/29/2021] [Accepted: 07/06/2021] [Indexed: 12/19/2022] Open
Abstract
Although persistent pain is estimated to affect about 20% of the adult population, current treatments have poor results. Polypharmacology, which is the administration of more than one drug targeting on two or more different sites of action, represents a prominent therapeutic approach for the clinical management of persistent pain. Thus, in the drug discovery process the "one-molecule-multiple targets" strategy nowadays is highly recognized. Indeed, multitarget ligands displaying a better antinociceptive activity with fewer side effects, combined with favorable pharmacokinetic and pharmacodynamic characteristics, have already been shown. Multitarget ligands possessing non-opioid/opioid and opioid/opioid mechanisms of action are considered as potential drug candidates for the management of various pain conditions. In particular, dual-target MOPr (mu opioid peptide receptor)/DOPr (delta opioid peptide receptor) ligands exhibit an improved antinociceptive profile associated with a reduced tolerance-inducing capability. The benzomorphan-based compounds LP1 and LP2 belong to this class of dual-target MOPr/DOPr ligands. In the present manuscript, the structure-activity relationships and the pharmacological fingerprint of LP1 and LP2 compounds as suitable drug candidates for persistent pain relief is described.
Collapse
Affiliation(s)
- Lorella Pasquinucci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Correspondence: (L.P.); (R.T.); Tel.: +39-095-738-4273 (L.P. & R.T.)
| | - Carmela Parenti
- Department of Drug and Health Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy;
| | - Zafiroula Georgoussi
- Laboratory of Cellular Signaling and Molecular Pharmacology, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos” Ag. Paraskevi-Attikis, 15310 Athens, Greece;
| | - Lorena Reina
- Postgraduate School of Clinical Pharmacology, Toxicology University of Catania, via S. Sofia n. 97, 95100 Catania, Italy;
| | - Emilia Tomarchio
- Postgraduate School of Anesthesiology and Intensive Care, University of Milan, Via Francesco Sforza, 35, 20122 Milan, Italy;
| | - Rita Turnaturi
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Correspondence: (L.P.); (R.T.); Tel.: +39-095-738-4273 (L.P. & R.T.)
| |
Collapse
|
10
|
Liu Q, He H, Mai L, Yang S, Fan W, Huang F. Peripherally Acting Opioids in Orofacial Pain. Front Neurosci 2021; 15:665445. [PMID: 34017236 PMCID: PMC8129166 DOI: 10.3389/fnins.2021.665445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
The activation of opioid receptors by exogenous or endogenous opioids can produce significant analgesic effects in peripheral tissues. Numerous researchers have demonstrated the expression of peripheral opioid receptors (PORs) and endogenous opioid peptides (EOPs) in the orofacial region. Growing evidence has shown the involvement of PORs and immune cell-derived EOPs in the modulation of orofacial pain. In this review, we discuss the role of PORs and EOPs in orofacial pain and the possible cellular mechanisms involved. Furthermore, the potential development of therapeutic strategies for orofacial pain is also summarized.
Collapse
Affiliation(s)
- Qing Liu
- Department of Pediatric Dentistry, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Department of Oral Anatomy and Physiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lijia Mai
- Department of Pediatric Dentistry, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Shengyan Yang
- Department of Pediatric Dentistry, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Wenguo Fan
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Fang Huang
- Department of Pediatric Dentistry, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
11
|
Ganesh CB, Vijayalaxmi. Neuroanatomical organization of methionine-enkephalinergic system in the brain of the Mozambique tilapia Oreochromis mossambicus. J Chem Neuroanat 2021; 115:101963. [PMID: 33957231 DOI: 10.1016/j.jchemneu.2021.101963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 11/24/2022]
Abstract
Enkephalins are a class of opioid peptides implicated in several physiological and neuroendocrine responses in vertebrates. In this study, using immunocytochemical or immunofluorescence technique, we examined the neuroanatomical distribution of methionine enkephalin (M-ENK) immunoreactivity in the central nervous system (CNS) of the cichlid fish Oreochromis mossambicus. In the telencephalon, no M-ENK-like-immunoreactive (M-ENK-L-ir) perikarya, but sparsely distributed fibres were detected in the glomerular layer and the granule cell layer of the olfactory bulb. Although intensely labeled M-ENK-L-ir cells and fibres were found in the pallium, no M-ENK immunoreactivity was observed in the subpallium. The preoptic area showed a few M-ENK-L-ir somata and dense innervations of fibres. In the hypothalamic area, M-ENK-L-ir cells and fibres were located in magnocellular and parvocellular subdivisions of the nucleus preopticus, and medial and lateral subdivisions of the nucleus lateralis tuberis. Surrounding the recessus lateralis of the third ventricle, several intensely stained and packed M-ENK-L-ir cells and fibres were seen in dorsal, lateral and ventral subdivisions of the nucleus recessus lateralis. In the diencephalon, M-ENK immunoreactivity was restricted to the habenula, the thalamus, the pretectal area and the nucleus posterior tuberis. Dense aggregations of M-ENK-L-ir fibres were seen in the mesencephalic subdivisions, the optic tectum and the torus semicircularis, whereas a few fusiform M-ENK-L-ir cells and fibres were scattered in the midbrain tegmentum. In the rhombencephalon, different populations of ovoid or spindle shaped M-ENK-L-ir cells were observed in the secondary gustatory nucleus, the sensory trigeminal nerve nucleus, the nucleus reticularis medialis and the vagal motor nucleus, whereas bands of fibres were seen in the rostral spinal cord. Collectively, the widespread distribution of M-ENK immunoreactivity in the CNS suggests a role for this opioid peptide in regulation of neuroendocrine control of reproduction and modulation of sensorimotor functions in fish.
Collapse
Affiliation(s)
- C B Ganesh
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad, 580 003, India.
| | - Vijayalaxmi
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad, 580 003, India
| |
Collapse
|
12
|
Recher M, Garabedian C, Aubry E, Sharma D, Butruille L, Storme L, De Jonckheere J. Opioid effect on the autonomic nervous system in a fetal sheep model. Arch Gynecol Obstet 2021; 304:73-80. [PMID: 33389095 DOI: 10.1007/s00404-020-05917-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/21/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE Opioid use during labour can interfere with cardiotocography patterns. Heart rate variability indirectly reflects a fluctuation in the autonomic nervous system and can be monitored through time and spectral analyses. This experimental study aimed to evaluate the impact of nalbuphine administration on the gasometric, cardiovascular, and autonomic nervous system responses in fetal sheep. METHODS This was an experimental study on chronically instrumented sheep fetuses (surgery at 128 ± 2 days of gestational age, term = 145 days). The model was based on a maternal intravenous bolus injection of nalbuphine, a semisynthetic opioid used as an analgesic during delivery. Fetal gasometric parameters (pH, pO2, pCO2, and lactates), hemodynamic parameters (fetal heart rate and mean arterial pressure), and autonomic nervous system tone (short-term and long-term variation, low-frequency domain, high-frequency domain, and fetal stress index) were recorded. Data obtained at 30-60 min after nalbuphine injection were compared to those recorded at baseline. RESULTS Eleven experiments were performed. Fetal heart rate, mean arterial pressure, and activities at low and high frequencies were stable after injection. Short-term variation decreased at T30 min (P = 0.02), and long-term variation decreased at T60 min (P = 0.02). Fetal stress index gradually increased and reached significance at T60 min (P = 0.02). Fetal gasometric parameters and lactate levels remained stable. CONCLUSION Maternal nalbuphine use during labour may lead to fetal heart changes that are caused by the effect of opioid on the autonomic nervous system; these fluctuations do not reflect acidosis.
Collapse
Affiliation(s)
- Morgan Recher
- ULR 2694, METRICS, Evaluation des Technologies de Santé et des Pratiques Médicales, University of Lille, 59000, Lille, France. .,Department of Paediatric Intensive Care Unit, CHU Lille, Jeanne de Flandre Hospital, 59000, Lille, France. .,Jeanne de Flandre Hospital, University of Lille Nord de France, 1 rue Eugène Avinée, 59037, Lille Cedex, France.
| | - Charles Garabedian
- ULR 2694, METRICS, Evaluation des Technologies de Santé et des Pratiques Médicales, University of Lille, 59000, Lille, France.,Department of Obstetrics, CHU Lille, Jeanne de Flandre Hospital, 59000, Lille, France
| | - Estelle Aubry
- ULR 2694, METRICS, Evaluation des Technologies de Santé et des Pratiques Médicales, University of Lille, 59000, Lille, France.,Department of Pediatric Surgery, CHU Lille, Jeanne de Flandre Hospital, 59000, Lille, France
| | - Dyuti Sharma
- ULR 2694, METRICS, Evaluation des Technologies de Santé et des Pratiques Médicales, University of Lille, 59000, Lille, France.,Department of Pediatric Surgery, CHU Lille, Jeanne de Flandre Hospital, 59000, Lille, France
| | - Laura Butruille
- ULR 2694, METRICS, Evaluation des Technologies de Santé et des Pratiques Médicales, University of Lille, 59000, Lille, France
| | - Laurent Storme
- ULR 2694, METRICS, Evaluation des Technologies de Santé et des Pratiques Médicales, University of Lille, 59000, Lille, France.,Department of Neonatology, CHU Lille, Jeanne de Flandre Hospital, 59000, Lille, France
| | - Julien De Jonckheere
- ULR 2694, METRICS, Evaluation des Technologies de Santé et des Pratiques Médicales, University of Lille, 59000, Lille, France.,CIC-IT 1403-biosensor and e-health, CHU Lille, 59000, Lille, France
| |
Collapse
|
13
|
Parker KE, Sugiarto E, Taylor AMW, Pradhan AA, Al-Hasani R. Pain, Motivation, Migraine, and the Microbiome: New Frontiers for Opioid Systems and Disease. Mol Pharmacol 2020; 98:433-444. [PMID: 32958571 PMCID: PMC7562975 DOI: 10.1124/mol.120.119438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022] Open
Abstract
For decades the broad role of opioids in addiction, neuropsychiatric disorders, and pain states has been somewhat well established. However, in recent years, with the rise of technological advances, not only is the existing dogma being challenged, but we are identifying new disease areas in which opioids play a critical role. This review highlights four new areas of exploration in the opioid field. The most recent addition to the opioid family, the nociceptin receptor system, shows promise as the missing link in understanding the neurocircuitry of motivation. It is well known that activation of the kappa opioid receptor system modulates negative affect and dysphoria, but recent studies now implicate the kappa opioid system in the modulation of negative affect associated with pain. Opioids are critical in pain management; however, the often-forgotten delta opioid receptor system has been identified as a novel therapeutic target for headache disorders and migraine. Lastly, changes to the gut microbiome have been shown to directly contribute to many of the symptoms of chronic opioid use and opioid related behaviors. This review summarizes the findings from each of these areas with an emphasis on identifying new therapeutic targets. SIGNIFICANCE STATEMENT: The focus of this minireview is to highlight new disease areas or new aspects of disease in which opioids have been implicated; this includes pain, motivation, migraine, and the microbiome. In some cases, this has resulted in the pursuit of a novel therapeutic target and resultant clinical trial. We believe this is very timely and will be a refreshing take on reading about opioids and disease.
Collapse
Affiliation(s)
- Kyle E Parker
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Elizabeth Sugiarto
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Anna M W Taylor
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Amynah A Pradhan
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Ream Al-Hasani
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| |
Collapse
|
14
|
Bertels Z, Pradhan AAA. Emerging Treatment Targets for Migraine and Other Headaches. Headache 2020; 59 Suppl 2:50-65. [PMID: 31291018 DOI: 10.1111/head.13585] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
Migraine is a complex disorder that is characterized by an assortment of neurological and systemic effects. While headache is the most prominent feature of migraine, a host of symptoms affecting many physiological functions are also observed before, during, and after an attack. Furthermore, migraineurs are heterogeneous and have a wide range of responses to migraine therapies. The recent approval of calcitonin gene-related-peptide based therapies has opened up the treatment of migraine and generated a renewed interest in migraine research and discovery. Ongoing advances in migraine research have identified a number of other promising therapeutic targets for this disorder. In this review, we highlight emergent treatments within the following biological systems: pituitary adenylate cyclase activating peptdie, 2 non-mu opioid receptors that have low abuse liability - the delta and kappa opioid receptors, orexin, and nitric oxide-based therapies. Multiple mechanisms have been identified in the induction and maintenance of migraine symptoms; and this divergent set of targets have highly distinct biological effects. Increasing the mechanistic diversity of the migraine tool box will lead to more treatment options and better patient care.
Collapse
Affiliation(s)
- Zachariah Bertels
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | | |
Collapse
|
15
|
Sakharkar AJ, Ganesh CB. Leucine-enkephalin-immunoreactive neurons in the brain of the cichlid fish Oreochromis mossambicus. Neuropeptides 2020; 81:101999. [PMID: 31843219 DOI: 10.1016/j.npep.2019.101999] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/26/2019] [Accepted: 12/09/2019] [Indexed: 12/29/2022]
Abstract
Enkephalins are the pentapeptides involved in pain relief and neuroendocrine responses with high affinity for delta opioid receptors in vertebrates. In the present investigation, we studied the distribution of leucine-enkephalin-immunoreactive (L-ENK-ir) neurons in the brain of the cichlid fish Oreochromis mossambicus. Application of the antisera against L-ENK revealed the presence of numerous L-ENK-ir perikarya and fibres in subdivisions of the dorsal and the ventral telencephalon, the medial olfactory tract and the nucleus entopeduncularis, whereas intensely labelled L-ENK-ir fibres were noticed in the olfactory bulb. Furthermore, the presence of L-ENK-ir cells and dense accumulations of fibres in the preoptic area and its subdivisions, the nucleus preopticus pars magnocellularis and the nucleus preopticus pars parvocellularis suggested a role for this peptide in regulation of reproduction. While intensely labelled cells and fibres were found in the nucleus lateralis tuberis pars lateralis as well as the nucleus lateralis tuberis pars medialis, some L-ENK-ir fibres were seen at the hypothalamo-hypophyseal tract indicating the possible hypophysiotrophic role for this peptide. Numerous L-ENK-ir cells and dense network of fibres were observed in the subdivisions of the nucleus recess lateralis and the pretectal area, whereas intensely labelled thick network of L-ENK- fibres were found in the ventromedial thalamic nucleus, the sub-layers of the optic tectum and the rostral spinal cord. The widespread distribution of L-ENK-immunoreactivity in the olfactory bulb, the telencephalon, the diencephalon and the mesencephalon regions of the brain as well as the spinal cord suggests the possible involvement of this peptide in the regulation of diverse functions such as neuroendocrine, antinociceptive, visual and olfactory responses in O. mossambicus.
Collapse
Affiliation(s)
- Amul J Sakharkar
- Department of Biotechnology, Savitribai Phule Pune University, Pune 411007, India
| | - C B Ganesh
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad 580 003, India.
| |
Collapse
|
16
|
Robins MT, Heinricher MM, Ryabinin AE. From Pleasure to Pain, and Back Again: The Intricate Relationship Between Alcohol and Nociception. Alcohol Alcohol 2020; 54:625-638. [PMID: 31509854 DOI: 10.1093/alcalc/agz067] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/09/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
AIMS A close and bidirectional relationship between alcohol consumption and pain has been previously reported and discussed in influential reviews. The goal of the present narrative review is to provide an update on the developments in this field in order to guide future research objectives. METHODS We evaluated both epidemiological and neurobiological literature interrogating the relationship between alcohol use and pain for the presence of significant effects. We outlined studies on interactions between alcohol use and pain using both self-reports and objective experimental measures and discussed potential underlying mechanisms of these interactions. RESULTS Epidemiological, preclinical and clinical literature point to three major interactions between alcohol use and pain: (a) alcohol use leading to hyperalgesia, (b) alcohol use moderating pain and hyperalgesia and (c) chronic pain as a risk factor predisposing to alcohol relapse. Neurobiological studies using animal models to assess these interactions have transitioned from mostly involuntary modes of experimenter-controlled alcohol administration to self-administration procedures, and increasingly indicate that neuronal circuits implicated in both withdrawal and anticipation stages of alcohol use disorder also have a role in chronic pain. Mechanistically, alterations in GABA, glutamate, the corticotropin-releasing factor system, endogenous opioids and protein kinase C appear to play crucial roles in this maladaptive overlap. CONCLUSIONS Many of the principles explaining the interactions between alcohol and pain remain on a strong foundation, but continuing progress in modeling these interactions and underlying systems will provide a clearer basis for understanding, and ultimately treating, the damaging aspects of this interaction.
Collapse
Affiliation(s)
- Meridith T Robins
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Mary M Heinricher
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.,Department of Neurological Surgery, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| |
Collapse
|
17
|
Modulation of the Negative Affective Dimension of Pain: Focus on Selected Neuropeptidergic System Contributions. Int J Mol Sci 2019; 20:ijms20164010. [PMID: 31426473 PMCID: PMC6720937 DOI: 10.3390/ijms20164010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/11/2022] Open
Abstract
It is well known that emotions can interfere with the perception of physical pain, as well as with the development and maintenance of painful conditions. On the other hand, somatic pain can have significant consequences on an individual’s affective behavior. Indeed, pain is defined as a complex and multidimensional experience, which includes both sensory and emotional components, thus exhibiting the features of a highly subjective experience. Over the years, neural pathways involved in the modulation of the different components of pain have been identified, indicating the existence of medial and lateral pain systems, which, respectively, project from medial or lateral thalamic nuclei to reach distinct cortex regions relating to specific functions. However, owing to the limited information concerning how mood state and painful input affect each other, pain treatment is frequently unsatisfactory. Different neuromodulators, including endogenous neuropeptides, appear to be involved in pain-related emotion and in its affective influence on pain perception, thus playing key roles in vulnerability and clinical outcome. Hence, this review article focuses on evidence concerning the modulation of the sensory and affective dimensions of pain, with particular attention given to some selected neuropeptidergic system contributions.
Collapse
|
18
|
DiCello JJ, Saito A, Rajasekhar P, Sebastian BW, McQuade RM, Gondin AB, Veldhuis NA, Canals M, Carbone SE, Poole DP. Agonist-dependent development of delta opioid receptor tolerance in the colon. Cell Mol Life Sci 2019; 76:3033-3050. [PMID: 30904952 PMCID: PMC11105391 DOI: 10.1007/s00018-019-03077-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/26/2019] [Accepted: 03/18/2019] [Indexed: 10/27/2022]
Abstract
The use of opioid analgesics is severely limited due to the development of intractable constipation, mediated through activation of mu opioid receptors (MOR) expressed by enteric neurons. The related delta opioid receptor (DOR) is an emerging therapeutic target for chronic pain, depression and anxiety. Whether DOR agonists also promote sustained inhibition of colonic transit is unknown. This study examined acute and chronic tolerance to SNC80 and ARM390, which were full and partial DOR agonists in neural pathways controlling colonic motility, respectively. Excitatory pathways developed acute and chronic tolerance to SNC80, whereas only chronic tolerance developed in inhibitory pathways. Both pathways remained functional after acute or chronic ARM390 exposure. Propagating colonic motor patterns were significantly reduced after acute or chronic SNC80 treatment, but not by ARM390 pre-treatment. These findings demonstrate that SNC80 has a prolonged inhibitory effect on propagating colonic motility. ARM390 had no effect on motor patterns and thus may have fewer gastrointestinal side-effects.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Benzamides/pharmacology
- Colon/drug effects
- Colon/physiology
- Drug Tolerance
- Electric Stimulation
- Mice
- Mice, Inbred C57BL
- Microscopy, Confocal
- Muscle Contraction/drug effects
- Neurons/metabolism
- Piperazines/pharmacology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Jesse J DiCello
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia.
| | - Ayame Saito
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Pradeep Rajasekhar
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Benjamin W Sebastian
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Rachel M McQuade
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Arisbel B Gondin
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Nicholas A Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Meritxell Canals
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Simona E Carbone
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Daniel P Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia.
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
19
|
Ellis CR, Racz R, Kruhlak NL, Kim MT, Hawkins EG, Strauss DG, Stavitskaya L. Assessing the Structural and Pharmacological Similarity of Newly Identified Drugs of Abuse to Controlled Substances Using Public Health Assessment via Structural Evaluation. Clin Pharmacol Ther 2019; 106:116-122. [PMID: 30957872 PMCID: PMC6617983 DOI: 10.1002/cpt.1418] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/16/2019] [Indexed: 12/17/2022]
Abstract
The US Food and Drug Administration's Center for Drug Evaluation and Research (CDER) developed an investigational Public Health Assessment via Structural Evaluation (PHASE) methodology to provide a structure-based evaluation of a newly identified opioid's risk to public safety. PHASE utilizes molecular structure to predict biological function. First, a similarity metric quantifies the structural similarity of a new drug relative to drugs currently controlled in the Controlled Substances Act (CSA). Next, software predictions provide the primary and secondary biological targets of the new drug. Finally, molecular docking estimates the binding affinity at the identified biological targets. The multicomponent computational approach coupled with expert review provides a rapid, systematic evaluation of a new drug in the absence of in vitro or in vivo data. The information provided by PHASE has the potential to inform law enforcement agencies with vital information regarding newly emerging illicit opioids.
Collapse
Affiliation(s)
- Christopher R. Ellis
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Rebecca Racz
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Naomi L. Kruhlak
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Marlene T. Kim
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Edward G. Hawkins
- Controlled Substances StaffCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - David G. Strauss
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Lidiya Stavitskaya
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
20
|
Moye LS, Tipton AF, Dripps I, Sheets Z, Crombie A, Violin JD, Pradhan AA. Delta opioid receptor agonists are effective for multiple types of headache disorders. Neuropharmacology 2019; 148:77-86. [PMID: 30553828 PMCID: PMC6467218 DOI: 10.1016/j.neuropharm.2018.12.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/19/2018] [Accepted: 12/12/2018] [Indexed: 12/20/2022]
Abstract
Headaches are highly disabling and are among the most common neurological disorders worldwide. Despite the high prevalence of headache, therapeutic options are limited. We recently identified the delta opioid receptor (DOR) as an emerging therapeutic target for migraine. In this study, we examined the effectiveness of a hallmark DOR agonist, SNC80, in disease models reflecting diverse headache disorders including: chronic migraine, post-traumatic headache (PTH), medication overuse headache by triptans (MOH), and opioid-induced hyperalgesia (OIH). To model chronic migraine C57BL/6J mice received chronic intermittent treatment with the known human migraine trigger, nitroglycerin. PTH was modeled by combining the closed head weight drop model with the nitroglycerin model of chronic migraine. For MOH and OIH, mice were chronically treated with sumatriptan or morphine, respectively. The development of periorbital and peripheral allodynia was observed in all four models; and SNC80 significantly inhibited allodynia in all cases. In addition, we also determined if chronic daily treatment with SNC80 would induce MOH/OIH, and we observed limited hyperalgesia relative to sumatriptan or morphine. Together, our results indicate that DOR agonists could be effective in multiple headache disorders, despite their distinct etiology, thus presenting a novel therapeutic target for headache.
Collapse
Affiliation(s)
- Laura S Moye
- Department of Psychiatry, University of Illinois at Chicago, USA
| | - Alycia F Tipton
- Department of Psychiatry, University of Illinois at Chicago, USA
| | - Isaac Dripps
- Department of Psychiatry, University of Illinois at Chicago, USA
| | - Zoie Sheets
- Department of Psychiatry, University of Illinois at Chicago, USA
| | | | | | - Amynah A Pradhan
- Department of Psychiatry, University of Illinois at Chicago, USA.
| |
Collapse
|
21
|
Cunningham CW, Elballa WM, Vold SU. Bifunctional opioid receptor ligands as novel analgesics. Neuropharmacology 2019; 151:195-207. [PMID: 30858102 DOI: 10.1016/j.neuropharm.2019.03.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/30/2019] [Accepted: 03/02/2019] [Indexed: 12/12/2022]
Abstract
Prolonged treatment of chronic severe pain with opioid analgesics is frought with problematic adverse effects including tolerance, dependence, and life-threatening respiratory depression. Though these effects are mediated predominately through preferential activation of μ opioid peptide (μOP) receptors, there is an emerging appreciation that actions at κOP and δOP receptors contribute to the observed pharmacologic and behavioral profile of μOP receptor agonists and may be targeted simultaneously to afford improved analgesic effects. Recent developments have also identified the related nociceptin opioid peptide (NOP) receptor as a key modulator of the effects of μOP receptor signaling. We review here the available literature describing OP neurotransmitter systems and highlight recent drug and probe design strategies.
Collapse
Affiliation(s)
| | - Waleed M Elballa
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, Mequon, WI, USA.
| | - Stephanie U Vold
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, Mequon, WI, USA.
| |
Collapse
|
22
|
DiCello JJ, Saito A, Rajasekhar P, Eriksson EM, McQuade RM, Nowell CJ, Sebastian BW, Fichna J, Veldhuis NA, Canals M, Bunnett NW, Carbone SE, Poole DP. Inflammation-associated changes in DOR expression and function in the mouse colon. Am J Physiol Gastrointest Liver Physiol 2018; 315:G544-G559. [PMID: 29927325 PMCID: PMC6230691 DOI: 10.1152/ajpgi.00025.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Endogenous opioids activate opioid receptors (ORs) in the enteric nervous system to control intestinal motility and secretion. The μ-OR mediates the deleterious side effects of opioid analgesics, including constipation, respiratory depression, and addiction. Although the δ-OR (DOR) is a promising target for analgesia, the function and regulation of DOR in the colon are poorly understood. This study provides evidence that endogenous opioids activate DOR in myenteric neurons that may regulate colonic motility. The DOR agonists DADLE, deltorphin II, and SNC80 inhibited electrically evoked contractions and induced neurogenic contractions in the mouse colon. Electrical, chemical, and mechanical stimulation of the colon evoked the release of endogenous opioids, which stimulated endocytosis of DOR in the soma and proximal neurites of myenteric neurons of transgenic mice expressing DOR fused to enhanced green fluorescent protein. In contrast, DOR was not internalized in nerve fibers within the circular muscle. Administration of dextran sulfate sodium induced acute colitis, which was accompanied by DOR endocytosis and an increased density of DOR-positive nerve fibers within the circular muscle. The potency with which SNC80 inhibited neurogenic contractions was significantly enhanced in the inflamed colon. This study demonstrates that DOR-expressing neurons in the mouse colon can be activated by exogenous and endogenous opioids. Activated DOR traffics to endosomes and inhibits neurogenic motility of the colon. DOR signaling is enhanced during intestinal inflammation. This study demonstrates functional expression of DOR by myenteric neurons and supports the therapeutic targeting of DOR in the enteric nervous system. NEW & NOTEWORTHY DOR is activated during physiologically relevant reflex stimulation. Agonist-evoked DOR endocytosis is spatially and temporally regulated. A significant proportion of DOR is internalized in myenteric neurons during inflammation. The relative proportion of all myenteric neurons that expressed DOR and the overlap with the nNOS-positive population are increased in inflammation. DOR-specific innervation of the circular muscle is increased in inflammation, and this is consistent with enhanced responsiveness to the DOR agonist SNC80.
Collapse
Affiliation(s)
- Jesse J. DiCello
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Ayame Saito
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Pradeep Rajasekhar
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Emily M. Eriksson
- 2Division of Population Health and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia,3Division of Infection and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia,4Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Rachel M. McQuade
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Cameron J. Nowell
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Benjamin W. Sebastian
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Jakub Fichna
- 5Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Nicholas A. Veldhuis
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,6Department of Genetics, University of Melbourne, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Meritxell Canals
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Nigel W. Bunnett
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia,8Department of Pharmacology and Therapeutics University of Melbourne, Parkville, Victoria, Australia,9Department of Surgery and Pharmacology, Columbia University, New York, New York
| | - Simona E. Carbone
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Daniel P. Poole
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia,10Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
23
|
Vicente-Sanchez A, Dripps IJ, Tipton AF, Akbari H, Akbari A, Jutkiewicz EM, Pradhan AA. Tolerance to high-internalizing δ opioid receptor agonist is critically mediated by arrestin 2. Br J Pharmacol 2018; 175:3050-3059. [PMID: 29722902 DOI: 10.1111/bph.14353] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/16/2018] [Accepted: 04/20/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Opioid δ receptor agonists are potent antihyperalgesics in chronic pain models, but tolerance develops after prolonged use. Previous evidence indicates that distinct forms of tolerance occur depending on the internalization properties of δ receptor agonists. As arrestins are important in receptor internalization, we investigated the role of arrestin 2 (β-arrestin 1) in mediating the development of tolerance induced by high- and low-internalizing δ receptor agonists. EXPERIMENTAL APPROACH We evaluated the effect of two δ receptor agonists with similar analgesic potencies, but either high-(SNC80) or low-(ARM390) internalization properties in wild-type (WT) and arrestin 2 knockout (KO) mice. We compared tolerance to the antihyperalgesic effects of these compounds in a model of inflammatory pain. We also examined tolerance to the convulsant effect of SNC80. Furthermore, effect of chronic treatment with SNC80 on δ agonist-stimulated [35 S]-GTPγS binding was determined in WT and KO mice. KEY RESULTS Arrestin 2 KO resulted in increased drug potency, duration of action and decreased acute tolerance to the antihyperalgesic effects of SNC80. In contrast, ARM390 produced similar effects in both WT and KO animals. Following chronic treatment, we found a marked decrease in the extent of tolerance to SNC80-induced antihyperalgesia and convulsions in arrestin 2 KO mice. Accordingly, δ receptors remained functionally coupled to G proteins in arrestin 2 KO mice chronically treated with SNC80. CONCLUSIONS AND IMPLICATIONS Overall, these results suggest that δ receptor agonists interact with arrestins in a ligand-specific manner, and tolerance to high- but not low-internalizing agonists are preferentially regulated by arrestin 2.
Collapse
Affiliation(s)
- Ana Vicente-Sanchez
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Isaac J Dripps
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Alycia F Tipton
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Heba Akbari
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Areeb Akbari
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Amynah A Pradhan
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Opioid use and abuse has led to a worldwide opioid epidemic. And while opioids are clinically useful when appropriately indicated, they are associated with a wide range of dangerous side effects and whether they are effective at treating or eliminating chronic pain is controversial. There has long been a need for the development of nonopioid alternative treatments for patients that live in pain, and until recently, only a few effective treatments were available. Today, there are a wide range of nonopioid treatments available including NSAIDs, acetaminophen, corticosteroids, nerve blocks, SSRIs, neurostimulators, and anticonvulsants. However, these treatments are still not entirely effective at treating pain, which has sparked a new exploration of novel nonopioid pharmacotherapies. RECENT FINDINGS This manuscript will outline the most recent trends in novel nonopioid pharmacotherapy development including tramadol/dexketoprofen, TrkA inhibitors, tapentadol, opioid agonists, Nektar 181, TRV 130, ßarrestin2, bisphosphonates, antibodies, sodium channel blockers, NMDA antagonists, TRP receptors, transdermal vitamin D, AAK1 kinase inhibition, calcitonin gene-related peptide (CGRP), TRPV4 antagonists, cholecystokinin, delta opioid receptor, neurokinin, and gene therapy. The pharmacotherapies discussed in this manuscript outline promising opioid alternatives which can change the future of chronic pain treatment.
Collapse
|
25
|
Abstract
Delta opioid receptors (δORs) regulate a number of physiological functions, and agonists for this receptor are being pursued for the treatment of mood disorders, chronic pain, and migraine. A major challenge to the development of these compounds is that, like many G-protein coupled receptors (GPCRs), agonists at the δOR can induce very different signaling and receptor trafficking events. This concept, known as ligand-directed signaling, functional selectivity, or biased agonism, can result in different agonists producing highly distinct behavioral consequences. In this chapter, we highlight the in vitro and in vivo evidence for ligand-directed signaling and trafficking at the δOR. A number of biological implications of agonist-directed signaling at the δOR have been demonstrated. Importantly, ligand-specific effects can impact both acute behavioral effects of delta agonists, as well as the long-term adaptations induced by chronic drug treatment. A better understanding of the specific signaling cascades that regulate these differential behavioral effects would help to guide rational drug design, ultimately resulting in δOR agonists with fewer adverse effects.
Collapse
Affiliation(s)
- Ana Vicente-Sanchez
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Amynah A Pradhan
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Psychiatry, UIC, 1601 W Taylor St (MC 912), Chicago, IL, 60612, USA.
| |
Collapse
|
26
|
Spahn V, Stein C. Targeting delta opioid receptors for pain treatment: drugs in phase I and II clinical development. Expert Opin Investig Drugs 2017; 26:155-160. [PMID: 28001096 DOI: 10.1080/13543784.2017.1275562] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Opioids are widely used to treat severe pain. Most clinically used opioids activate µ-opioid receptors (MOR). Their ligands induce potent analgesia but also adverse effects. The δ-opioid receptor (DOR) is another member of the opioid receptor family that has been under intense investigation with the aim to avoid MOR-induced side effects. Areas covered: This article reviews DOR ligands which appeared to be promising after preclinical evaluation. A literature search using Pubmed, Cochrane library, ClinicalTrials.gov, EudraCT, AdisInsight database and EBSCO Online Library was conducted. Out of numerous newly synthesized molecules, only few candidates entered phase I and/or II clinical investigation. The publicly accessible results are presented here. Expert opinion: Many compounds showed potent DOR-specific pain inhibition in preclinical studies. ADL5859 and ADL5747 entered clinical trials and successfully passed phase I. However, in phase II studies the primary endpoint (pain reduction) was not met and further investigation was terminated. A third compound, NP2, is in phase II clinical evaluation and results are pending. These findings suggest a potential of DOR ligands according to preclinical studies. Further clinical research and secondary analysis of unpublished data is needed to identify molecules which are useful in humans.
Collapse
Affiliation(s)
- Viola Spahn
- a Klinik für Anästhesiologie und operative Intensivmedizin , Charité Campus Benjamin Franklin, Freie Universität Berlin , Berlin , Germany
| | - Christoph Stein
- a Klinik für Anästhesiologie und operative Intensivmedizin , Charité Campus Benjamin Franklin, Freie Universität Berlin , Berlin , Germany.,b Multifunctional Biomaterials for Medicine , Helmholtz Virtual Institute , Teltow , Germany
| |
Collapse
|
27
|
Rice FL, Xie JY, Albrecht PJ, Acker E, Bourgeois J, Navratilova E, Dodick DW, Porreca F. Anatomy and immunochemical characterization of the non-arterial peptidergic diffuse dural innervation of the rat and Rhesus monkey: Implications for functional regulation and treatment in migraine. Cephalalgia 2016; 37:1350-1372. [DOI: 10.1177/0333102416677051] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Objective The interplay between neuronal innervation and other cell types underlies the physiological functions of the dura mater and contributes to pathophysiological conditions such as migraine. We characterized the extensive, but understudied, non-arterial diffuse dural innervation (DDI) of the rat and Rhesus monkey. Methods We used a comprehensive integrated multi-molecular immunofluorescence labeling strategy to extensively profile the rat DDI and to a lesser extent that of the Rhesus monkey. Results The DDI was distributed across a dense, pervasive capillary network and included free nerve endings of peptidergic CGRP-expressing C fibers that were closely intertwined with noradrenergic (NA) sympathetic fibers and thin-caliber nonpeptidergic “C/Aδ” fibers. These newly identified C/Aδ fibers were unmyelinated, like C fibers, but expressed NF200, usually indicative of Aδ fibers, and uniquely co-labeled for the CGRP co-receptor, RAMP1. Slightly-larger caliber NF200-positive fibers co-labeled for myelin basic protein (MBP) and terminated as unbranched corpuscular endings. The DDI peptidergic fibers co-labeled for the lectin IB4 and expressed presumably excitatory α1-adrenergic receptors, as well as inhibitory 5HT1D receptors and the delta opioid receptor (δOR), but rarely the mu opioid receptor (µOR). Labeling for P2X3, TRPV1, TRPA1, and parasympathetic markers was not observed in the DDI. Interpretation These results suggest potential functional interactions, wherein peptidergic DDI fibers may be activated by stress-related sympathetic activity, resulting in CGRP release that could be detected in the circulation. CGRP may also activate nonpeptidergic C/Aδ fibers that are likely mechanosensitive or polymodal, leading to activation of post-synaptic pain transmission circuits. The distribution of α1-adrenergic receptors, RAMP1, and the unique expression of the δOR on CGRP-expressing DDI fibers suggest strategies for functional modulation and application to therapy.
Collapse
Affiliation(s)
- Frank L Rice
- Integrated Tissue Dynamics LLC, Rensselaer, NY, USA
| | - Jennifer Y Xie
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | | | - Emily Acker
- Integrated Tissue Dynamics LLC, Rensselaer, NY, USA
| | | | - Edita Navratilova
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | - David W Dodick
- Departments of Collaborative Research and Neurology, Mayo Clinic, Scottsdale, AZ, USA
| | - Frank Porreca
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
- Departments of Collaborative Research and Neurology, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|