1
|
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
|
2
|
Turnaturi R, Piana S, Spoto S, Costanzo G, Reina L, Pasquinucci L, Parenti C. From Plant to Chemistry: Sources of Active Opioid Antinociceptive Principles for Medicinal Chemistry and Drug Design. Molecules 2023; 28:7089. [PMID: 37894567 PMCID: PMC10609244 DOI: 10.3390/molecules28207089] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/28/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Pain continues to be an enormous global health challenge, with millions of new untreated or inadequately treated patients reported annually. With respect to current clinical applications, opioids remain the mainstay for the treatment of pain, although they are often associated with serious side effects. To optimize their tolerability profiles, medicinal chemistry continues to study novel ligands and innovative approaches. Among them, natural products are known to be a rich source of lead compounds for drug discovery, and they hold potential for pain management. Traditional medicine has had a long history in clinical practice due to the fact that nature provides a rich source of active principles. For instance, opium had been used for pain management until the 19th century when its individual components, such as morphine, were purified and identified. In this review article, we conducted a literature survey aimed at identifying natural products interacting either directly with opioid receptors or indirectly through other mechanisms controlling opioid receptor signaling, whose structures could be interesting from a drug design perspective.
Collapse
Affiliation(s)
- Rita Turnaturi
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy;
| | - Silvia Piana
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy;
| | - Salvatore Spoto
- Department of Drug and Health Sciences, Section of Pharmacology and Toxicology, University of Catania, 95125 Catania, Italy; (S.S.); (C.P.)
| | - Giuliana Costanzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy;
| | - Lorena Reina
- Postgraduate School of Clinical Pharmacology, Toxicology University of Catania, Via Santa Sofia n. 97, 95100 Catania, Italy;
| | - Lorella Pasquinucci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy;
| | - Carmela Parenti
- Department of Drug and Health Sciences, Section of Pharmacology and Toxicology, University of Catania, 95125 Catania, Italy; (S.S.); (C.P.)
| |
Collapse
|
3
|
Puls K, Wolber G. Solving an Old Puzzle: Elucidation and Evaluation of the Binding Mode of Salvinorin A at the Kappa Opioid Receptor. Molecules 2023; 28:718. [PMID: 36677775 PMCID: PMC9861206 DOI: 10.3390/molecules28020718] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/07/2022] [Accepted: 12/16/2022] [Indexed: 01/13/2023] Open
Abstract
The natural product Salvinorin A (SalA) was the first nitrogen-lacking agonist discovered for the opioid receptors and exhibits high selectivity for the kappa opioid receptor (KOR) turning SalA into a promising analgesic to overcome the current opioid crisis. Since SalA's suffers from poor pharmacokinetic properties, particularly the absence of gastrointestinal bioavailability, fast metabolic inactivation, and subsequent short duration of action, the rational design of new tailored analogs with improved clinical usability is highly desired. Despite being known for decades, the binding mode of SalA within the KOR remains elusive as several conflicting binding modes of SalA were proposed hindering the rational design of new analgesics. In this study, we rationally determined the binding mode of SalA to the active state KOR by in silico experiments (docking, molecular dynamics simulations, dynophores) in the context of all available mutagenesis studies and structure-activity relationship (SAR) data. To the best of our knowledge, this is the first comprehensive evaluation of SalA's binding mode since the determination of the active state KOR crystal structure. SalA binds above the morphinan binding site with its furan pointing toward the intracellular core while the C2-acetoxy group is oriented toward the extracellular loop 2 (ECL2). SalA is solely stabilized within the binding pocket by hydrogen bonds (C210ECL2, Y3127.35, Y3137.36) and hydrophobic contacts (V1182.63, I1393.33, I2946.55, I3167.39). With the disruption of this interaction pattern or the establishment of additional interactions within the binding site, we were able to rationalize the experimental data for selected analogs. We surmise the C2-substituent interactions as important for SalA and its analogs to be experimentally active, albeit with moderate frequency within MD simulations of SalA. We further identified the non-conserved residues 2.63, 7.35, and 7.36 responsible for the KOR subtype selectivity of SalA. We are confident that the elucidation of the SalA binding mode will promote the understanding of KOR activation and facilitate the development of novel analgesics that are urgently needed.
Collapse
Affiliation(s)
| | - Gerhard Wolber
- Department of Biology, Chemistry and Pharmacy, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| |
Collapse
|
4
|
Critical interactions between opioid and cannabinoid receptors during tolerance and physical dependence development to opioids in the murine gastrointestinal tract: proof of concept. Pharmacol Rep 2021; 73:1147-1154. [PMID: 34133018 PMCID: PMC8413198 DOI: 10.1007/s43440-021-00291-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/20/2021] [Accepted: 05/31/2021] [Indexed: 11/05/2022]
Abstract
Introduction Tolerance (TOL) and physical dependence (PD) constitute important limitations of opioid therapy. The aim of our study was to validate research tools to investigate TOL and PD and to characterize the interactions between opioid (OR) and cannabinoid (CB) receptors in these processes in the GI tract. Methods TOL was assessed through the comparison of morphine ability to inhibit electrically evoked smooth muscles contractility in the mouse ileum that was previously incubated with/without morphine for 1 h. To evaluate the PD, the ileum was incubated with morphine for 10 min, then challenged with naloxone to induce withdrawal response (WR). The OR/CB interactions were evaluated using mixed agonist (PR-38) and AM-251 (CB1 antagonist). Results The inhibitory effect of morphine on ileal contractions was weaker in tissue incubated with this opioid than in tissue incubated without opioid. The opposite was noted for PR-38. In tissues exposed to morphine, but not to PR-38, naloxone induced a WR. The blockage of CB1 receptors with AM-251 before the addition of PR-38 resulted in a naloxone-induced WR. Conclusion The co-activation of OR and CB reduced development of TOL and PD to opioids in the mouse GI tract and mixed OR/CB agonists are promising alternative to currently used opioid drugs.
Collapse
|
5
|
Chakraborty S, Majumdar S. Natural Products for the Treatment of Pain: Chemistry and Pharmacology of Salvinorin A, Mitragynine, and Collybolide. Biochemistry 2021; 60:1381-1400. [PMID: 32930582 PMCID: PMC7982354 DOI: 10.1021/acs.biochem.0c00629] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pain remains a very pervasive problem throughout medicine. Classical pain management is achieved through the use of opiates belonging to the mu opioid receptor (MOR) class, which have significant side effects that hinder their utility. Pharmacologists have been trying to develop opioids devoid of side effects since the isolation of morphine from papaver somniferum, more commonly known as opium by Sertürner in 1804. The natural products salvinorin A, mitragynine, and collybolide represent three nonmorphinan natural product-based targets, which are potent selective agonists of opioid receptors, and emerging next-generation analgesics. In this work, we review the phytochemistry and medicinal chemistry efforts on these templates and their effects on affinity, selectivity, analgesic actions, and a myriad of other opioid-receptor-related behavioral effects.
Collapse
Affiliation(s)
- Soumen Chakraborty
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of Medicine, St. Louis, Missouri 63110, United States; Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Susruta Majumdar
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of Medicine, St. Louis, Missouri 63110, United States; Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
6
|
Crowley RS, Riley AP, Alder AF, Anderson RJ, Luo D, Kaska S, Maynez P, Kivell BM, Prisinzano TE. Synthetic Studies of Neoclerodane Diterpenes from Salvia divinorum: Design, Synthesis, and Evaluation of Analogues with Improved Potency and G-protein Activation Bias at the μ-Opioid Receptor. ACS Chem Neurosci 2020; 11:1781-1790. [PMID: 32383854 DOI: 10.1021/acschemneuro.0c00191] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Previous structure-activity relationship (SAR) studies identified the first centrally acting, non-nitrogenous μ-opioid receptor (MOR) agonist, kurkinorin (1), derived from salvinorin A. In an effort to further probe the physiological effects induced upon activation of MORs with this nonmorphine scaffold, a variety of analogues were synthesized and evaluated in vitro for their ability to activate G-proteins and recruit β-arrestin-2 upon MOR activation. Through these studies, compounds that are potent agonists at MORs and either biased toward β-arrestin-2 recruitment or biased toward G-protein activation have been identified. One such compound, 25, has potent activity and selectivity at the MOR over KOR with bias for G-protein activation. Impressively, 25 is over 100× more potent than morphine and over 5× more potent than fentanyl in vitro and elicits antinociception with limited tolerance development in vivo. This is especially significant given that 25 lacks a basic nitrogen and other ionizable groups present in other opioid ligand classes.
Collapse
Affiliation(s)
- Rachel S. Crowley
- Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas 66047, United States
| | - Andrew P. Riley
- Department of Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Amy F. Alder
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Richard J. Anderson
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Dan Luo
- Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas 66047, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Sophia Kaska
- Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas 66047, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Pamela Maynez
- Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas 66047, United States
| | - Bronwyn M. Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Thomas E. Prisinzano
- Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas 66047, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| |
Collapse
|
7
|
Luo K, Bao Y, Liu F, Xiao C, Li K, Zhang C, Huang R, Lin J, Zhang J, Jin Y. Synthesis and biological evaluation of novel benzylidene-succinimide derivatives as noncytotoxic antiangiogenic inhibitors with anticolorectal cancer activity in vivo. Eur J Med Chem 2019; 179:805-827. [DOI: 10.1016/j.ejmech.2019.06.094] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/05/2019] [Accepted: 06/30/2019] [Indexed: 02/07/2023]
|
8
|
Keasling AW, Pandey P, Doerksen RJ, Pedrino GR, Costa EA, da Cunha LC, Zjawiony JK, Fajemiroye JO. Salvindolin elicits opioid system-mediated antinociceptive and antidepressant-like activities. J Psychopharmacol 2019; 33:865-881. [PMID: 31192780 DOI: 10.1177/0269881119849821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Salvinorin A is known as a highly selective kappa opioid receptor agonist with antinociceptive but mostly pro-depressive effects. AIMS In this article, we present its new semisynthetic analog with preferential mu opioid affinity, and promising antinociceptive, as well as antidepressant-like activities. METHODS Competitive binding studies were performed for salvindolin with kappa opioid and mu opioid. The mouse model of nociception (acetic-acid-induced writhing, formalin, and hot plate tests), depression (forced swim and tail suspension tests), and the open field test, were used to evaluate antinociceptive, antidepressant-like, and locomotion effects, respectively, of salvindolin. We built a 3-D molecular model of the kappa opioid receptor, using a mu opioid X-ray crystal structure as a template, and docked salvindolin into the two proteins. RESULTS/OUTCOMES Salvindolin showed affinity towards kappa opioid and mu opioid receptors but with 100-fold mu opioid preference. Tests of salvindolin in mice revealed good oral bioavailability, antinociceptive, and antidepressive-like effects, without locomotor incoordination. Docking of salvindolin showed strong interactions with the mu opioid receptor which matched well with experimental binding data. Salvindolin-induced behavioral changes in the hot plate and forced swim tests were attenuated by naloxone (nonselective opioid receptor antagonist) and/or naloxonazine (selective mu opioid receptor antagonist) but not by nor-binaltorphimine (selective kappa opioid receptor antagonist). In addition, WAY100635 (a selective serotonin 1A receptor antagonist) blocked the antidepressant-like effect of salvindolin. CONCLUSIONS/INTERPRETATION By simple chemical modification, we were able to modulate the pharmacological profile of salvinorin A, a highly selective kappa opioid receptor agonist, to salvindolin, a ligand with preferential mu opioid receptor affinity and activity on the serotonin 1A receptor. With its significant antinociceptive and antidepressive-like activities, salvindolin has the potential to be an analgesic and/or antidepressant drug candidate.
Collapse
Affiliation(s)
- Adam W Keasling
- 1 Department of BioMolecular Sciences, Division of Pharmacognosy, University of Mississippi, University, MS, USA.,2 Department of BioMolecular Sciences, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS, USA
| | - Pankaj Pandey
- 3 Department of BioMolecular Sciences, Division of Medicinal Chemistry, University of Mississippi, University, MS, USA
| | - Robert J Doerksen
- 2 Department of BioMolecular Sciences, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS, USA.,3 Department of BioMolecular Sciences, Division of Medicinal Chemistry, University of Mississippi, University, MS, USA
| | - Gustavo R Pedrino
- 4 Department of Physiology, Federal University of Goiás, Goiânia, Brazil
| | - Elson A Costa
- 5 Department of Pharmacology, Federal University of Goiás, Goiânia, Brazil
| | - Luiz C da Cunha
- 6 Center for Studies and Toxicological-Pharmacological Research, Federal University of Goiás, Goiânia, Brazil
| | - Jordan K Zjawiony
- 1 Department of BioMolecular Sciences, Division of Pharmacognosy, University of Mississippi, University, MS, USA.,2 Department of BioMolecular Sciences, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS, USA
| | - James O Fajemiroye
- 5 Department of Pharmacology, Federal University of Goiás, Goiânia, Brazil.,6 Center for Studies and Toxicological-Pharmacological Research, Federal University of Goiás, Goiânia, Brazil.,7 Department of Pharmaceutical Science, University Center of Anápolis - Unievangélica, Anápolis, Brazil
| |
Collapse
|
9
|
Zjawiony JK, Machado AS, Menegatti R, Ghedini PC, Costa EA, Pedrino GR, Lukas SE, Franco OL, Silva ON, Fajemiroye JO. Cutting-Edge Search for Safer Opioid Pain Relief: Retrospective Review of Salvinorin A and Its Analogs. Front Psychiatry 2019; 10:157. [PMID: 30971961 PMCID: PMC6445891 DOI: 10.3389/fpsyt.2019.00157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 03/04/2019] [Indexed: 12/21/2022] Open
Abstract
Over the years, pain has contributed to low life quality, poor health, and economic loss. Opioids are very effective analgesic drugs for treating mild, moderate, or severe pain. Therapeutic application of opioids has been limited by short and long-term side effects. These side effects and opioid-overuse crisis has intensified interest in the search for new molecular targets and drugs. The present review focuses on salvinorin A and its analogs with the aim of exploring their structural and pharmacological profiles as clues for the development of safer analgesics. Ethnopharmacological reports and growing preclinical data have demonstrated the antinociceptive effect of salvinorin A and some of its analogs. The pharmacology of analogs modified at C-2 dominates the literature when compared to the ones from other positions. The distinctive binding affinity of these analogs seems to correlate with their chemical structure and in vivo antinociceptive effects. The high susceptibility of salvinorin A to chemical modification makes it an important pharmacological tool for cellular probing and developing analogs with promising analgesic effects. Additional research is still needed to draw reliable conclusions on the therapeutic potential of salvinorin A and its analogs.
Collapse
Affiliation(s)
- Jordan K Zjawiony
- Division of Pharmacognosy, Department of BioMolecular Sciences, School of Pharmacy, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS, United States
| | - Antônio S Machado
- Laboratory of Medicinal Pharmaceutical Chemistry, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, Brazil
| | - Ricardo Menegatti
- Laboratory of Medicinal Pharmaceutical Chemistry, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, Brazil
| | - Paulo C Ghedini
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Goiás, Goiânia, Brazil
| | - Elson A Costa
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Goiás, Goiânia, Brazil
| | - Gustavo R Pedrino
- Department of Physiology, Universidade Federal de Goiás, Goiânia, Brazil
| | - Scott E Lukas
- McLean Imaging Center, Harvard Medical School, McLean Hospital, Belmont, MA, United States
| | - Octávio L Franco
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil.,Programa de Pós-graduação em Patologia Molecular, Universidade de Brasília, Brasília, Brazil
| | - Osmar N Silva
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - James O Fajemiroye
- Department of Physiology, Universidade Federal de Goiás, Goiânia, Brazil.,Centro Universitário de Anápolis, Unievangélica, Anápolis, Brazil
| |
Collapse
|
10
|
Roach JJ, Shenvi RA. A review of salvinorin analogs and their kappa-opioid receptor activity. Bioorg Med Chem Lett 2018; 28:1436-1445. [PMID: 29615341 PMCID: PMC5912166 DOI: 10.1016/j.bmcl.2018.03.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 12/23/2022]
Abstract
The plant metabolite salvinorin A potently and selectively agonizes the human kappa-opioid receptor, an emerging target for next-generation analgesics. Here we review analogs of the salvinorin chemotype and their effects on selectivity, affinity and potency. Extensive peripheral modifications using isolated salvinorin A have delivered a trove of SAR information. More deep-seated changes are now possible by advances in chemical synthesis.
Collapse
Affiliation(s)
- Jeremy J Roach
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ryan A Shenvi
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
11
|
Wareing B, Urbisch D, Kolle SN, Honarvar N, Sauer UG, Mehling A, Landsiedel R. Prediction of skin sensitization potency sub-categories using peptide reactivity data. Toxicol In Vitro 2017; 45:134-145. [DOI: 10.1016/j.tiv.2017.08.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 06/11/2017] [Accepted: 08/21/2017] [Indexed: 12/28/2022]
|
12
|
Allosteric modulation model of the mu opioid receptor by herkinorin, a potent not alkaloidal agonist. J Comput Aided Mol Des 2017; 31:467-482. [PMID: 28364251 DOI: 10.1007/s10822-017-0016-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/10/2017] [Indexed: 10/19/2022]
Abstract
Modulation of opioid receptors is the primary choice for pain management and structural information studies have gained new horizons with the recently available X-ray crystal structures. Herkinorin is one of the most remarkable salvinorin A derivative with high affinity for the mu opioid receptor, moderate selectivity and lack of nitrogen atoms on its structure. Surprisingly, binding models for herkinorin are lacking. In this work, we explore binding models of herkinorin using automated docking, molecular dynamics simulations, free energy calculations and available experimental information. Our herkinorin D-ICM-1 binding model predicted a binding free energy of -11.52 ± 1.14 kcal mol-1 by alchemical free energy estimations, which is close to the experimental values -10.91 ± 0.2 and -10.80 ± 0.05 kcal mol-1 and is in agreement with experimental structural information. Specifically, D-ICM-1 molecular dynamics simulations showed a water-mediated interaction between D-ICM-1 and the amino acid H2976.52, this interaction coincides with the co-crystallized ligands. Another relevant interaction, with N1272.63, allowed to rationalize herkinorin's selectivity to mu over delta opioid receptors. Our suggested binding model for herkinorin is in agreement with this and additional experimental data. The most remarkable observation derived from our D-ICM-1 model is that herkinorin reaches an allosteric sodium ion binding site near N1503.35. Key interactions in that region appear relevant for the lack of β-arrestin recruitment by herkinorin. This interaction is key for downstream signaling pathways involved in the development of side effects, such as tolerance. Future SAR studies and medicinal chemistry efforts will benefit from the structural information presented in this work.
Collapse
|
13
|
Fajemiroye JO, Prabhakar PR, Cunha LCD, Costa EA, Zjawiony JK. 22-azidosalvinorin A exhibits antidepressant-like effect in mice. Eur J Pharmacol 2017; 800:96-106. [PMID: 28219707 DOI: 10.1016/j.ejphar.2017.02.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 10/20/2022]
Abstract
The increasing cases of depression has made the searches for new drugs and understanding of the underligning neurobiology of this psychiatric disorder a necessity. Here, we modified the structure of salvinorin A (a known halucinogen) and investigated antidepressant-like activity of its four derivatives; 22-methylsulfanylsalvinorin A(SA1), 2-O-cinnamoylsalvinorin B (CSB), 22-azidosalvinorin A (SA2), and 2-O-(4'-azidophenylsulfonyl)salvinorin B (SA3). Prior to behavioural tests (Irwin test, open field test - OFT, forced swimming test - FST and tail suspension test - TST), SA1 was prepared by reacting salvinorin B and methylthioacetic acid with 89% yield; CSB was obtained from the reaction of salvinorin B and cinnamic acid with 92% yield; SA2 was obtained from the reaction of salvinorin B and azidoacetic acid with 81% yield; and SA3 was prepared by reacting salvinorin B with 4-azidophenylsulfonyl chloride with 80% yield. Oral treatment of mice with these derivatives (1-1000mg/kg) did not elicit toxic sign or death. Unlike SA, SA1, CSB and SA3, treatment with SA2 (5, 10 and 20mg/kg) decreased the immobility (TST and FST) and swimming time (FST) without altering locomotor activity in OFT. A decrease in the immobility time in TST and FST confirmed antidepressant-like property of SA2. Although p-chlorophenylalanine (serotonin depletor) or WAY100635 (selective 5-HT1A receptor antagonist) did not attenuate effect of SA2, alpha-methyl-para-tyrosine (catecholamine depletor) and prazosin (selective α1-receptor antagonist) attenuated this effect. SA2 mildly inhibited monoamine oxidase and showed affinity for α1A, α1B, α1D and κ-opioid receptor subtypes. In summary, SA2 induced monoamine-mediated antidepressant-like effect.
Collapse
Affiliation(s)
- James Oluwagbamigbe Fajemiroye
- Department of Pharmacology, Federal University of Goias, Campus Samambaia, 74001-970 Goiania, GO, Brazil; Center for Studies and Toxicological-Pharmacological Research, Faculty of Pharmacy, Federal University of Goiás, PMB 131, CEP 74001-970, Goiânia, Brazil.
| | - Polepally Reddy Prabhakar
- Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, P.O. Box 1848, MS 38677, USA
| | - Luiz Carlos da Cunha
- Center for Studies and Toxicological-Pharmacological Research, Faculty of Pharmacy, Federal University of Goiás, PMB 131, CEP 74001-970, Goiânia, Brazil
| | - Elson Alves Costa
- Department of Pharmacology, Federal University of Goias, Campus Samambaia, 74001-970 Goiania, GO, Brazil
| | - Jordan K Zjawiony
- Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, P.O. Box 1848, MS 38677, USA
| |
Collapse
|
14
|
Abstract
This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants). This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
15
|
Bartuzi D, Kaczor AA, Matosiuk D. Activation and Allosteric Modulation of Human μ Opioid Receptor in Molecular Dynamics. J Chem Inf Model 2015; 55:2421-34. [DOI: 10.1021/acs.jcim.5b00280] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Damian Bartuzi
- Department
of Synthesis and Chemical Technology of Pharmaceutical Substances
with Computer Modeling Lab, Faculty of Pharmacy with Division of Medical
Analytics, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland
| | - Agnieszka A. Kaczor
- Department
of Synthesis and Chemical Technology of Pharmaceutical Substances
with Computer Modeling Lab, Faculty of Pharmacy with Division of Medical
Analytics, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland
- School
of Pharmacy, University of Eastern Finland, Yliopistonranta 1, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Dariusz Matosiuk
- Department
of Synthesis and Chemical Technology of Pharmaceutical Substances
with Computer Modeling Lab, Faculty of Pharmacy with Division of Medical
Analytics, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland
| |
Collapse
|
16
|
Salaga M, Polepally PR, Zielinska M, Marynowski M, Fabisiak A, Murawska N, Sobczak K, Sacharczuk M, Do Rego JC, Roth BL, Zjawiony JK, Fichna J. Salvinorin A analogues PR-37 and PR-38 attenuate compound 48/80-induced itch responses in mice. Br J Pharmacol 2015; 172:4331-41. [PMID: 26040667 DOI: 10.1111/bph.13212] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 05/26/2015] [Accepted: 06/01/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The opioid system plays a crucial role in several physiological processes in the CNS and in the periphery. It has also been shown that selective opioid receptor agonists exert potent inhibitory action on pruritus and pain. In this study we examined whether two analogues of Salvinorin A, PR-37 and PR-38, exhibit antipruritic properties in mice. EXPERIMENTAL APPROACH To examine the antiscratch effect of PR-37 and PR-38 we used a mouse model of compound 48/80-induced pruritus. In order to elucidate the mechanism of action of tested compounds, specific antagonists of opioid and cannabinoid receptors were used. The effect of PR-37 on the CNS was assessed by measuring motor parameters and exploratory behaviours in mice. KEY RESULTS PR-37 and PR-38, jnjected s.c., significantly reduced the number of compound 48/80-induced scratching behaviours in mice in a dose- and time-dependent manner. PR-38 was also active when orally administered. The antiscratch activity of PR-37 was blocked by the selective κ opioid receptor antagonist, nor-binaltorphimine, and that of PR-38 by the selective μ opioid receptor antagonist, β-funaltrexamine. CONCLUSION AND IMPLICATIONS In conclusion, a novel framework for the development of new antipruritic drugs derived from salvinorin A has been validated.
Collapse
Affiliation(s)
- M Salaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - P R Polepally
- Department of BioMolecular Sciences, Division of Pharmacognosy and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, USA
| | - M Zielinska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - M Marynowski
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - A Fabisiak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - N Murawska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - K Sobczak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - M Sacharczuk
- Department of Molecular Cytogenetic, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzebiec, Poland
| | - J C Do Rego
- Platform of Behavioural Analysis (SCAC), Institute for Research and Innovation in Biomedicine (IRIB), Faculty of Medicine & Pharmacy, University of Rouen, Rouen Cedex, France
| | - B L Roth
- Department of Pharmacology, Division of Chemical Biology and Medicinal Chemistry, Medical School, NIMH Psychoactive Drug Screening Program, University of North Carolina, Chapel Hill, NC, USA
| | - J K Zjawiony
- Department of BioMolecular Sciences, Division of Pharmacognosy and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, USA
| | - J Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
17
|
Nieto CT, Gonzalez-Nunez V, Rodríguez RE, Diez D, Garrido NM. Design, synthesis, pharmacological evaluation and molecular dynamics of β-amino acids morphan-derivatives as novel ligands for opioid receptors. Eur J Med Chem 2015; 101:150-62. [PMID: 26134550 DOI: 10.1016/j.ejmech.2015.06.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 06/06/2015] [Accepted: 06/09/2015] [Indexed: 10/23/2022]
Abstract
Structure-Activity Relationship (SAR) is a current approach in the design of new pharmacological agents. We previously reported the synthesis of a novel analogue of morphine, a 2-azabicyclo[3.3.1]nonane, which contains a β-amino acid. This bicyclic core exhibits two distinctive chemical handles for further elaboration, which allowed us to create a library of morphan-containing compounds by in silico molecular docking on the μ opioid receptor. Lead candidates were synthesized and biological tests were performed to evaluate their ability to bind to opioid receptors. The four top compounds, three phenyl esters and an N-phenylethyl morphan derivative, were selected for Molecular Dynamics simulations to get topological and thermodynamic information. Aromatic morphan derivatives displayed an interacting domain which fits into a hydrophobic cleft and the effect of the substituents in their affinity was explained by the differences in the calculated binding free energies. Our results indicate that the 3D arrangement of the aromatic ring in the morphine derivatives is not a key issue for a specific ligand - μ receptor interaction. Thus, these morphan derivatives represent a new class of opioid receptor ligands which may be of great use in the clinical practice.
Collapse
Affiliation(s)
- Carlos T Nieto
- Departamento de Química Orgánica, Faculty of Chemistry, Universidad de Salamanca, Spain
| | - Veronica Gonzalez-Nunez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Neuroscience Institute of Castilla y Leon (INCyL), University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), Spain
| | - Raquel E Rodríguez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Neuroscience Institute of Castilla y Leon (INCyL), University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), Spain
| | - David Diez
- Departamento de Química Orgánica, Faculty of Chemistry, Universidad de Salamanca, Spain
| | - Narciso M Garrido
- Departamento de Química Orgánica, Faculty of Chemistry, Universidad de Salamanca, Spain.
| |
Collapse
|
18
|
Shang Y, Filizola M. Opioid receptors: Structural and mechanistic insights into pharmacology and signaling. Eur J Pharmacol 2015; 763:206-13. [PMID: 25981301 DOI: 10.1016/j.ejphar.2015.05.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 03/02/2015] [Accepted: 05/11/2015] [Indexed: 01/18/2023]
Abstract
Opioid receptors are important drug targets for pain management, addiction, and mood disorders. Although substantial research on these important subtypes of G protein-coupled receptors has been conducted over the past two decades to discover ligands with higher specificity and diminished side effects, currently used opioid therapeutics remain suboptimal. Luckily, recent advances in structural biology of opioid receptors provide unprecedented insights into opioid receptor pharmacology and signaling. We review here a few recent studies that have used the crystal structures of opioid receptors as a basis for revealing mechanistic details of signal transduction mediated by these receptors, and for the purpose of drug discovery.
Collapse
Affiliation(s)
- Yi Shang
- Icahn School of Medicine at Mount Sinai, Department of Structural and Chemical Biology, One Gustave, L. Levy Place, Box 1677, New York, NY 10029, USA
| | - Marta Filizola
- Icahn School of Medicine at Mount Sinai, Department of Structural and Chemical Biology, One Gustave, L. Levy Place, Box 1677, New York, NY 10029, USA.
| |
Collapse
|
19
|
Guerrieri E, Mallareddy JR, Tóth G, Schmidhammer H, Spetea M. Synthesis and pharmacological evaluation of [(3)H]HS665, a novel, highly selective radioligand for the kappa opioid receptor. ACS Chem Neurosci 2015; 6:456-63. [PMID: 25496417 DOI: 10.1021/cn5002792] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Herein we report the radiolabeling and pharmacological investigation of a novel radioligand, the N-cyclobutylmethyl substituted diphenethylamine [(3)H]HS665, designed to bind selectively to the kappa opioid peptide (KOP) receptor, a target of therapeutic interest for the treatment of a variety of human disorders (i.e., pain, affective disorders, drug addiction, and psychotic disorders). HS665 was prepared in tritium-labeled form by a dehalotritiated method resulting in a specific activity of 30.65 Ci/mmol. Radioligand binding studies were performed to establish binding properties of [(3)H]HS665 to the recombinant human KOP receptor in membranes from Chinese hamster ovary cells stably expressing human KOP receptors (CHOhKOP) and to the native neuronal KOP receptor in guinea pig brain membranes. Binding of [(3)H]HS665 was specific and saturable in both tissue preparations. A single population of high affinity binding sites was labeled by [(3)H]HS665 in membranes from CHOhKOP cells and guinea pig brain with similar equilibrium dissociation constants, Kd, 0.45 and 0.64 nM, respectively. Average receptor density of [(3)H]HS665 recognition sites were 5564 and 154 fmol/mg protein in CHOhKOP cells and guinea pig brain, respectively. This study shows that the new radioligand distinguishes and labels KOP receptors specifically in neuronal and cellular systems expressing KOP receptors, making this molecule a valuable tool in probing structural and functional mechanisms governing ligand-KOP receptor interactions in both a recombinant and native in vitro setting.
Collapse
Affiliation(s)
- Elena Guerrieri
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Jayapal Reddy Mallareddy
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvari krt 62, H-6726 Szeged, Hungary
| | - Géza Tóth
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvari krt 62, H-6726 Szeged, Hungary
| | - Helmut Schmidhammer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| |
Collapse
|
20
|
Riley AP, Groer CE, Young D, Ewald AW, Kivell BM, Prisinzano TE. Synthesis and κ-opioid receptor activity of furan-substituted salvinorin A analogues. J Med Chem 2014; 57:10464-75. [PMID: 25426797 PMCID: PMC4281103 DOI: 10.1021/jm501521d] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The neoclerodane diterpene salvinorin A, found in the leaves of Salvia divinorum, is a potent κ-opioid receptor agonist, making it an attractive scaffold for development into a treatment for substance abuse. Although several successful semisynthetic studies have been performed to elucidate structure-activity relationships, the lack of analogues with substitutions to the furan ring of salvinorin A has prevented a thorough understanding of its role in binding to the κ-opioid receptor. Herein we report the synthesis of several salvinorin A derivatives with modified furan rings. Evaluation of these compounds in a functional assay indicated that sterically less demanding substitutions are preferred, suggesting the furan ring is bound in a congested portion of the binding pocket. The most potent of the analogues successfully reduced drug-seeking behavior in an animal model of drug-relapse without producing the sedation observed with other κ-opioid agonists.
Collapse
Affiliation(s)
- Andrew P Riley
- Department of Chemistry and ‡Department of Medicinal Chemistry, School of Pharmacy, The University of Kansas , Lawrence, Kansas 66045, United States
| | | | | | | | | | | |
Collapse
|