1
|
Liu W, Wang D, Wang L, Hu S, Jiang Y, Wang Y, Cai X, Chen J. Receptor dimers and biased ligands: Novel strategies for targeting G protein-coupled receptors. Pharmacol Ther 2025; 269:108829. [PMID: 40023322 DOI: 10.1016/j.pharmthera.2025.108829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/16/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
G protein-coupled receptors (GPCRs) are the largest superfamily of membrane receptors. They regulate physiological and pathological processes such as metabolic homeostasis, cell proliferation and differentiation, and the immune response, and are one of the most important classes of drug targets, being targeted by 30-40 % of marketed drugs. A growing number of studies continue to reveal the complexity of GPCRs, especially their ability to interact with each other to form higher-order structures such as homodimers and heterodimers, which have different functions than monomers, and are involved in disease development and progression. The existence of GPCR homodimers and heterodimers is opening up new directions in drug discovery and development to harness their therapeutic potential. Particularly striking is the ability of GPCR dimers to trigger unique biased signalling pathways, which exquisitely balance the relationship between therapeutic effects and side effects. By suppressing adverse reactions and enhancing beneficial drug effects, GPCR dimers provide an unprecedented opportunity to minimise side effects, maximise therapeutic efficacy and enhance safety. This review aims to highlight the latest research advances in GPCR dimerization and GPCR-biased signalling, focusing on the development of dimer-targeting and biased ligands as innovative drugs that will likely provide new strategies for treating GPCR-related diseases as well as a better understanding of drug design for compounds that target GPCRs. GPCRs will play an increasingly important role in precision medicine and personalised therapy, leading us towards a safer, more efficient and smarter pharmaceutical future.
Collapse
Affiliation(s)
- Wenkai Liu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, 261042, PR China
| | - Dexiu Wang
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, 261042, PR China
| | - Luoqi Wang
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, 261042, PR China
| | - Shujuan Hu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, 261042, PR China
| | - Yunlu Jiang
- Neurobiology Institute, Jining Medical University, Jining, Shandong 272067, PR China
| | - Yixiang Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong 261042, PR China
| | - Xin Cai
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, 261042, PR China.
| | - Jing Chen
- Neurobiology Institute, Jining Medical University, Jining, Shandong 272067, PR China; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
2
|
Childers W, Elokely K, Abou-Gharbia M. Dezocine and Addiction: Friend or Foe? Pharmaceuticals (Basel) 2025; 18:386. [PMID: 40143162 PMCID: PMC11946148 DOI: 10.3390/ph18030386] [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: 02/11/2025] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
The neurological effects of opium were first described over 8000 years ago. Morphine was isolated in 1803 and by the mid-1800s had become both a pain-relieving blessing and an addictive curse. As part of the crusade to identify safer and more reliable alternatives to morphine, dezocine (Dalgan®) was marketed in the US in 1986. Its use was discontinued in the US in 2011 without revealing the reasons, but it remains one of the most widely used analgesic agents in China today. Dezocine's unique pharmacology makes it an effective analgesic with limited opioid-associated side effects and little or no reported potential for dependence and addiction. In addition, dezocine's blocking effect on serotonin and norepinephrine transporters recommends its further exploration as a potential treatment for various chronic and neuropathic pain conditions. Most recently, data suggest that dezocine might represent a viable treatment for addiction management. This report focuses on the data supporting dezocine's non-addictive profile and its potential use to treat opioid addiction and withdrawal, as well as recent efforts to generate formulations of dezocine that support sub-chronic and chronic dosing.
Collapse
Affiliation(s)
- Wayne Childers
- Department of Pharmaceutical Sciences, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA;
| | - Khaled Elokely
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, WY 82071, USA;
| | - Magid Abou-Gharbia
- Department of Pharmaceutical Sciences, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA;
| |
Collapse
|
3
|
Zangrandi L, Fogli B, Mutti A, Staritzbichler R, Most V, Hildebrand PW, Heilbronn R, Schwarzer C. Structure-function relationship of dynorphin B variants using naturally occurring amino acid substitutions. Front Pharmacol 2024; 15:1484730. [PMID: 39539623 PMCID: PMC11557314 DOI: 10.3389/fphar.2024.1484730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Dynorphins (Dyn) represent the subset of endogenous opioid peptides with the highest binding affinity to kappa opioid receptors (KOPrs). Activation of the G-protein-coupled pathway of KOPrs has strong anticonvulsant effects. Dyn also bind to mu (MOPrs) and delta opioid receptors (DOPrs) with lower affinity and can activate the β-arrestin pathway. To fully exploit the therapeutic potential of dynorphins and reduce potential unwanted effects, increased selectivity for KOPrs combined with reduced activation of the mTOR complex would be favorable. Therefore, we investigated a series of dynorphin B (DynB) variants, substituted in one or two positions with naturally occurring amino acids for differential opioid receptor activation, applying competitive radio binding assays, GTPγS assays, PRESTO-Tango, and Western blotting on single-opioid receptor-expressing cells. Seven DynB derivatives displayed at least 10-fold increased selectivity for KOPrs over either MOPrs or DOPrs. The highest selectivity for KOPrs over MOPrs was obtained with DynB_G3M/Q8H, and the highest selectivity for KOPrs over DOPrs was obtained with DynB_L5S. Increased selectivity for KOPr over MOPr and DOPr was based on a loss of affinity or potency at MOPr and DOPr rather than a higher affinity or potency at KOPr. This suggests that the investigated amino acid exchanges in positions 3, 5, and 8 are of higher importance for binding and activation of MOPr or DOPr than of KOPr. In tests for signal transduction using the GTPγS assay, none of the DynB derivatives displayed increased potency. The three tested variants with substitutions of glycine to methionine in position 3 displayed reduced efficacy and are, therefore, considered partial agonists. The two most promising activating candidates were further investigated for functional selectivity between the G-protein and the β-arrestin pathway, as well as for activation of mTOR. No difference was detected in the respective read-outs, compared to wild-type DynB. Our data indicate that the assessment of affinity to KOPr alone is not sufficient to predict either potency or efficacy of peptidergic agonists on KOPr. Further assessment of downstream pathways is required to allow more reliable predictions of in vivo effects.
Collapse
Affiliation(s)
- Luca Zangrandi
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
- Clinic for Neurology and Experimental Neurology, AG Gene Therapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Barbara Fogli
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Mutti
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - René Staritzbichler
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Victoria Most
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Peter W. Hildebrand
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Berlin, Germany
| | - Regine Heilbronn
- Clinic for Neurology and Experimental Neurology, AG Gene Therapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christoph Schwarzer
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
Damiescu R, Elbadawi M, Dawood M, Klauck SM, Bringmann G, Efferth T. Aniquinazoline B, a Fungal Natural Product, Activates the μ-Opioid Receptor. ChemMedChem 2024; 19:e202400213. [PMID: 38781501 DOI: 10.1002/cmdc.202400213] [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: 04/18/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024]
Abstract
The development of new μ-opioid receptor (MOR) agonists without the undesirable side effects, such as addiction or respiratory depression, has been a difficult challenge over the years. In the search for new compounds, we screened our chemical database of over 40.000 substances and further assessed the best 100 through molecular docking. We selected the top 10 compounds and evaluated them for their biological activity and potential to influence cyclic adenosine monophosphate (cAMP) levels. From the tested compounds, compound 7, called aniquinazoline B, belonging to the quinazolinone alkaloids class and isolated from the marine fungus Aspergillus nidulans, showed promising results, by inhibiting cAMP levels and in vitro binding to MOR, verified through microscale thermophoresis. Transcriptomic data investigation profiled the genes affected by compound 7 and discovered activation of different pathways compared to opioids. The western blot analysis revealed compound 7 as a balanced ligand, activating both p-ERK1/2 and β-arrestin1/2 pathways, showing this is a favorable candidate to be further tested.
Collapse
Affiliation(s)
- Roxana Damiescu
- Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, Germany
| | - Mohamed Elbadawi
- Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, Germany
| | - Mona Dawood
- Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, Germany
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ) Heidelberg, National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ, University Hospital Heidelberg, Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Thomas Efferth
- Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, Germany
| |
Collapse
|
5
|
Fan YZ, Duan YL, Chen CT, Wang Y, Zhu AP. Advances in attenuating opioid-induced respiratory depression: A narrative review. Medicine (Baltimore) 2024; 103:e38837. [PMID: 39029082 PMCID: PMC11398798 DOI: 10.1097/md.0000000000038837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
Opioids exert analgesic effects by agonizing opioid receptors and activating signaling pathways coupled to receptors such as G-protein and/or β-arrestin. Concomitant respiratory depression (RD) is a common clinical problem, and improvement of RD is usually achieved with specific antagonists such as naloxone; however, naloxone antagonizes opioid analgesia and may produce more unknown adverse effects. In recent years, researchers have used various methods to isolate opioid receptor-mediated analgesia and RD, with the aim of preserving opioid analgesia while attenuating RD. At present, the focus is mainly on the development of new opioids with weak respiratory inhibition or the use of non-opioid drugs to stimulate breathing. This review reports recent advances in novel opioid agents, such as mixed opioid receptor agonists, peripheral selective opioid receptor agonists, opioid receptor splice variant agonists, biased opioid receptor agonists, and allosteric modulators of opioid receptors, as well as in non-opioid agents, such as AMPA receptor modulators, 5-hydroxytryptamine receptor agonists, phosphodiesterase-4 inhibitors, and nicotinic acetylcholine receptor agonists.
Collapse
Affiliation(s)
- Yong-Zheng Fan
- The 991st Hospital of Joint Logistic Support Force of People's Liberation Army, Xiangyang, China
| | - Yun-Li Duan
- Xiangyang No. 4 Middle School Compulsory Education Department, Xiangyang, China
| | - Chuan-Tao Chen
- Taihe Country People's Hospital·The Taihe Hospital of Wannan Medical College, Fuyang, China
| | - Yu Wang
- The 991st Hospital of Joint Logistic Support Force of People's Liberation Army, Xiangyang, China
| | - An-Ping Zhu
- The 991st Hospital of Joint Logistic Support Force of People's Liberation Army, Xiangyang, China
| |
Collapse
|
6
|
Barakat A, Munro G, Heegaard AM. Finding new analgesics: Computational pharmacology faces drug discovery challenges. Biochem Pharmacol 2024; 222:116091. [PMID: 38412924 DOI: 10.1016/j.bcp.2024.116091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/10/2024] [Accepted: 02/23/2024] [Indexed: 02/29/2024]
Abstract
Despite the worldwide prevalence and huge burden of pain, pain is an undertreated phenomenon. Currently used analgesics have several limitations regarding their efficacy and safety. The discovery of analgesics possessing a novel mechanism of action has faced multiple challenges, including a limited understanding of biological processes underpinning pain and analgesia and poor animal-to-human translation. Computational pharmacology is currently employed to face these challenges. In this review, we discuss the theory, methods, and applications of computational pharmacology in pain research. Computational pharmacology encompasses a wide variety of theoretical concepts and practical methodological approaches, with the overall aim of gaining biological insight through data acquisition and analysis. Data are acquired from patients or animal models with pain or analgesic treatment, at different levels of biological organization (molecular, cellular, physiological, and behavioral). Distinct methodological algorithms can then be used to analyze and integrate data. This helps to facilitate the identification of biological molecules and processes associated with pain phenotype, build quantitative models of pain signaling, and extract translatable features between humans and animals. However, computational pharmacology has several limitations, and its predictions can provide false positive and negative findings. Therefore, computational predictions are required to be validated experimentally before drawing solid conclusions. In this review, we discuss several case study examples of combining and integrating computational tools with experimental pain research tools to meet drug discovery challenges.
Collapse
Affiliation(s)
- Ahmed Barakat
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, Egypt.
| | | | - Anne-Marie Heegaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Hennessy MR, Gutridge AM, French AR, Rhoda ES, Meqbil YJ, Gill M, Kashyap Y, Appourchaux K, Paul B, Wang ZJ, van Rijn RM, Riley AP. Modified Akuamma Alkaloids with Increased Potency at the Mu-opioid Receptor. J Med Chem 2023; 66:3312-3326. [PMID: 36827198 PMCID: PMC10037270 DOI: 10.1021/acs.jmedchem.2c01707] [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] [Indexed: 02/25/2023]
Abstract
Akuammine (1) and pseudoakuammigine (2) are indole alkaloids found in the seeds of the akuamma tree (Picralima nitida). Both alkaloids are weak agonists of the mu opioid receptor (μOR); however, they produce minimal effects in animal models of antinociception. To probe the interactions of 1 and 2 at the opioid receptors, we have prepared a collection of 22 semisynthetic derivatives. Evaluation of this collection at the μOR and kappa opioid receptor (κOR) revealed structural-activity relationship trends and derivatives with improved potency at the μOR. Most notably, the introduction of a phenethyl moiety to the N1 of 2 produces a 70-fold increase in potency and a 7-fold increase in selectivity for the μOR. The in vitro potency of this compound resulted in increased efficacy in the tail-flick and hot-plate assays of antinociception. The improved potency of these derivatives highlights the promise of exploring natural product scaffolds to probe the opioid receptors.
Collapse
Affiliation(s)
- Madeline R Hennessy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612 USA
| | - Anna M Gutridge
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907 USA
| | - Alexander R French
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907 USA
- Departments of Neurology and Bioengineering, University of Illinois Chicago, Chicago, IL 60612 USA
| | - Elizabeth S Rhoda
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907 USA
| | - Yazan J. Meqbil
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907 USA
| | - Meghna Gill
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612 USA
| | - Yavnika Kashyap
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612 USA
| | - Kevin Appourchaux
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Barnali Paul
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Zaijie Jim Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612 USA
- Departments of Neurology and Bioengineering, University of Illinois Chicago, Chicago, IL 60612 USA
| | - Richard M. van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907 USA
- Purdue Institute for Drug Discovery, West Lafayette, IN 47907 USA
- Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907 USA
- Purdue Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, IN 47907 USA
| | - Andrew P. Riley
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612 USA
| |
Collapse
|
8
|
Inhibitory Effects of a Novel μ-Opioid Receptor Nonpeptide Antagonist, UD-030, on Morphine-Induced Conditioned Place Preference. Int J Mol Sci 2023; 24:ijms24043351. [PMID: 36834763 PMCID: PMC9961271 DOI: 10.3390/ijms24043351] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Although opioids are widely used to treat moderate to severe pain, opioid addiction and the opioid overdose epidemic are becoming more serious. Although opioid receptor antagonists/partial agonists, such as naltrexone and buprenorphine, have relatively low selectivity for the μ-opioid receptor (MOP), they have been used for the management of opioid use disorder. The utility of highly selective MOP antagonists remains to be evaluated. Here, we biologically and pharmacologically evaluated a novel nonpeptide ligand, UD-030, as a selective MOP antagonist. UD-030 had more than 100-fold higher binding affinity for the human MOP (Ki = 3.1 nM) than for δ-opioid, κ-opioid, and nociceptin receptors (Ki = 1800, 460, and 1800 nM, respectively) in competitive binding assays. The [35S]-GTPγS binding assay showed that UD-030 acts as a selective MOP full antagonist. The oral administration of UD-030 dose-dependently suppressed the acquisition and expression of morphine-induced conditioned place preference in C57BL/6J mice, and its effects were comparable to naltrexone. These results indicate the UD-030 may be a new candidate for the treatment of opioid use disorder, with characteristics that differ from traditional medications that are in clinical use.
Collapse
|
9
|
Zhao Y, Ren X, Li F, Jia B, Wang D, Jia H, Jiao X, Wang L, Li J. P2Y1 receptor in the colonic submucosa of rats and its association with opioid‑induced constipation. Exp Ther Med 2022; 25:67. [PMID: 36605532 PMCID: PMC9798462 DOI: 10.3892/etm.2022.11766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 11/03/2022] [Indexed: 12/15/2022] Open
Abstract
The aim of the present study was to explore the expression changes of P2Y purinergic receptor 1 (P2Y1) in the distal colonic submucosa of opioid-induced constipation (OIC) rats and its association with the occurrence of OIC, an OIC rat model was generated by intraperitoneal injection of loperamide hydrochloride, a selective agonist of µ-opioid receptors (MORs). At 7 days post-treatment, the model was assessed by analyzing stool scores and calculating the gastrointestinal (GI) transit ratio of rats. The distribution of P2Y1-expressing neurons in the colonic submucosal plexus was demonstrated by immunofluorescence (IF). Western blotting was performed to evaluate the expression changes of MOR, P2Y1 and ATP synthase subunit β (ATPB) proteins in the colonic submucosa, while reverse transcription-quantitative PCR (RT-qPCR) analysis was performed to determine the relative mRNA expression of MOR and P2Y1. After 7 days, the feces of OIC rats exhibited an appearance of sausage-shaped pieces and both the stool weight and GI transit ratio of OIC rats were significantly decreased. IF revealed co-expression of P2Y1 and calbindin and MOR and ATPB in the nerve cells of the distal colonic submucosal plexus. Moreover, RT-qPCR analysis showed that the MOR mRNA levels were significantly increased in the distal colonic submucosa of OIC rats, while mRNA levels of P2Y1 were decreased. WB showed that in the distal colonic submucosa of OIC rats, MOR protein expression was increased, whereas that of P2Y1 was significantly decreased. GI transit ratio analysis suggested that the P2Y agonist ATP significantly relieved constipation symptoms in rats, while the P2Y inhibitor MRS2179 aggravated these symptoms. Finally, P2Y1 expression change was shown to be associated with the occurrence of OIC, while expression of MOR and P2Y1 was associated with OIC development in rats.
Collapse
Affiliation(s)
- Yuqiong Zhao
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Xiaojie Ren
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Fan Li
- Department of Gastrointestinal Surgery, Xiantao First People's Hospital Affiliated to Yangtze University, Xiantao, Hubei 433000, P.R. China
| | - Binghan Jia
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Dengke Wang
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Hua Jia
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Xuwen Jiao
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Lixin Wang
- The Medical Laboratory Center of General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China,Correspondence to: Dr Junping Li, Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Junping Li
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China,Correspondence to: Dr Junping Li, Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| |
Collapse
|
10
|
Lee YS. Peptidomimetics and Their Applications for Opioid Peptide Drug Discovery. Biomolecules 2022; 12:biom12091241. [PMID: 36139079 PMCID: PMC9496382 DOI: 10.3390/biom12091241] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Despite various advantages, opioid peptides have been limited in their therapeutic uses due to the main drawbacks in metabolic stability, blood-brain barrier permeability, and bioavailability. Therefore, extensive studies have focused on overcoming the problems and optimizing the therapeutic potential. Currently, numerous peptide-based drugs are being marketed thanks to new synthetic strategies for optimizing metabolism and alternative routes of administration. This tutorial review briefly introduces the history and role of natural opioid peptides and highlights the key findings on their structure-activity relationships for the opioid receptors. It discusses details on opioid peptidomimetics applied to develop therapeutic candidates for the treatment of pain from the pharmacological and structural points of view. The main focus is the current status of various mimetic tools and the successful applications summarized in tables and figures.
Collapse
Affiliation(s)
- Yeon Sun Lee
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
11
|
Zhang J, Song C, Dai J, Li L, Yang X, Chen Z. Mechanism of opioid addiction and its intervention therapy: Focusing on the reward circuitry and mu-opioid receptor. MedComm (Beijing) 2022; 3:e148. [PMID: 35774845 PMCID: PMC9218544 DOI: 10.1002/mco2.148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/09/2022] Open
Abstract
Opioid abuse and addiction have become a global pandemic, posing tremendous health and social burdens. The rewarding effects and the occurrence of withdrawal symptoms are the two mainstays of opioid addiction. Mu-opioid receptors (MORs), a member of opioid receptors, play important roles in opioid addiction, mediating both the rewarding effects of opioids and opioid withdrawal syndrome (OWS). The underlying mechanism of MOR-mediated opioid rewarding effects and withdrawal syndrome is of vital importance to understand the nature of opioid addiction and also provides theoretical basis for targeting MORs to treat drug addiction. In this review, we first briefly introduce the basic concepts of MORs, including their structure, distribution in the nervous system, endogenous ligands, and functional characteristics. We focused on the brain circuitry and molecular mechanism of MORs-mediated opioid reward and withdrawal. The neuroanatomical and functional elements of the neural circuitry of the reward system underlying opioid addiction were thoroughly discussed, and the roles of MOR within the reward circuitry were also elaborated. Furthermore, we interrogated the roles of MORs in OWS, along with the structural basis and molecular adaptions of MORs-mediated withdrawal syndrome. Finally, current treatment strategies for opioid addiction targeting MORs were also presented.
Collapse
Affiliation(s)
- Jia‐Jia Zhang
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Chang‐Geng Song
- Department of NeurologyXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Ji‐Min Dai
- Department of Hepatobiliary SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Ling Li
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Xiang‐Min Yang
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Zhi‐Nan Chen
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
12
|
Koekkoek LL, van der Gun LL, Serlie MJ, la Fleur SE. The Clash of Two Epidemics: the Relationship Between Opioids and Glucose Metabolism. Curr Diab Rep 2022; 22:301-310. [PMID: 35593927 PMCID: PMC9188528 DOI: 10.1007/s11892-022-01473-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW We are currently in the midst of a global opioid epidemic. Opioids affect many physiological processes, but one side effect that is not often taken into consideration is the opioid-induced alteration in blood glucose levels. RECENT FINDINGS This review shows that the vast majority of studies report that opioid stimulation increases blood glucose levels. In addition, plasma levels of the endogenous opioid β-endorphin rise in response to low blood glucose. In contrast, in hyperglycaemic baseline conditions such as in patients with type 2 diabetes mellitus (T2DM), opioid stimulation lowers blood glucose levels. Furthermore, obesity itself alters sensitivity to opioids, changes opioid receptor expression and increases plasma β-endorphin levels. Thus, opioid stimulation can have various side effects on glycaemia that should be taken into consideration upon prescribing opioid-based medication, and more research is needed to unravel the interaction between obesity, glycaemia and opioid use.
Collapse
Affiliation(s)
- Laura L Koekkoek
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, Amsterdam, Netherlands
- Department of Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, K2-283, 1105 AZ, Amsterdam, the Netherlands
- Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, Netherlands
| | - Luna L van der Gun
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, Amsterdam, Netherlands
- Department of Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, K2-283, 1105 AZ, Amsterdam, the Netherlands
- Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, K2-283, 1105 AZ, Amsterdam, the Netherlands
| | - Susanne E la Fleur
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, Amsterdam, Netherlands.
- Department of Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, K2-283, 1105 AZ, Amsterdam, the Netherlands.
- Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, Netherlands.
| |
Collapse
|
13
|
Rouault AAJ, Buscaglia P, Sebag JA. MRAP2 inhibits β-arrestin recruitment to the ghrelin receptor by preventing GHSR1a phosphorylation. J Biol Chem 2022; 298:102057. [PMID: 35605660 PMCID: PMC9190059 DOI: 10.1016/j.jbc.2022.102057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 11/19/2022] Open
Abstract
The melanocortin receptor accessory protein 2 (MRAP2) is essential for several physiological functions of the ghrelin receptor growth hormone secretagogue receptor 1a (GHSR1a), including increasing appetite and suppressing insulin secretion. In the absence of MRAP2, GHSR1a displays high constitutive activity and a weak G-protein-mediated response to ghrelin and readily recruits β-arrestin. In the presence of MRAP2, however, G-protein-mediated signaling via GHSR1a is strongly dependent on ghrelin stimulation and the recruitment of β-arrestin is significantly diminished. To better understand how MRAP2 modifies GHSR1a signaling, here we investigated the role of several phosphorylation sites within the C-terminal tail and third intracellular loop of GHSR1a, as well as the mechanism behind MRAP2-mediated inhibition of β-arrestin recruitment. We show that Ser252 and Thr261 in the third intracellular loop of GHSR1a contribute to β-arrestin recruitment, whereas the C-terminal region is not essential for β-arrestin interaction. Additionally, we found that MRAP2 inhibits GHSR1a phosphorylation by blocking the interaction of GRK2 and PKC with the receptor. Taken together, these data suggest that MRAP2 alters GHSR1a signaling by directly impacting the phosphorylation state of the receptor and that the C-terminal tail of GHSR1a prevents rather than contribute to β-arrestin recruitment.
Collapse
Affiliation(s)
- Alix A J Rouault
- Department of Molecular Physiology and Biophysics, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA; F.O.E.D.R.C, Iowa City, Iowa, USA; Pappajohn Biomedical Institute, Iowa City, Iowa, USA; Iowa Neuroscience Institute, Iowa City, Iowa, USA
| | - Paul Buscaglia
- Department of Molecular Physiology and Biophysics, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA; F.O.E.D.R.C, Iowa City, Iowa, USA; Pappajohn Biomedical Institute, Iowa City, Iowa, USA; Iowa Neuroscience Institute, Iowa City, Iowa, USA
| | - Julien A Sebag
- Department of Molecular Physiology and Biophysics, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA; F.O.E.D.R.C, Iowa City, Iowa, USA; Pappajohn Biomedical Institute, Iowa City, Iowa, USA; Iowa Neuroscience Institute, Iowa City, Iowa, USA.
| |
Collapse
|
14
|
Wang X, Bao C, Li Z, Yue L, Hu L. Side Effects of Opioids Are Ameliorated by Regulating TRPV1 Receptors. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19042387. [PMID: 35206575 PMCID: PMC8872563 DOI: 10.3390/ijerph19042387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/06/2022] [Accepted: 02/09/2022] [Indexed: 11/23/2022]
Abstract
Humans have used opioids to suppress moderate to severe pain for thousands of years. However, the long-term use of opioids has several adverse effects, such as opioid tolerance, opioid-induced hyperalgesia, and addiction. In addition, the low efficiency of opioids in controlling neuropathic pain limits their clinical applications. Combining nonopioid analgesics with opioids to target multiple sites along the nociceptive pathway may alleviate the side effects of opioids. This study reviews the feasibility of reducing opioid side effects by regulating the transient receptor potential vanilloid 1 (TRPV1) receptors and summarizes the possible underlying mechanisms. Blocking and activating TRPV1 receptors can improve the therapeutic profile of opioids in different manners. TRPV1 and μ-opioid receptors are bidirectionally regulated by β-arrestin2. Thus, drug combinations or developing dual-acting drugs simultaneously targeting μ-opioid and TRPV1 receptors may mitigate opioid tolerance and opioid-induced hyperalgesia. In addition, TRPV1 receptors, especially expressed in the dorsal striatum and nucleus accumbens, participate in mediating opioid reward, and its regulation can reduce the risk of opioid-induced addiction. Finally, co-administration of TRPV1 antagonists and opioids in the primary action sites of the periphery can significantly relieve neuropathic pain. In general, the regulation of TRPV1 may potentially ameliorate the side effects of opioids and enhance their analgesic efficacy in neuropathic pain.
Collapse
Affiliation(s)
- Xiaqing Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; (X.W.); (C.B.); (Z.L.)
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chongyu Bao
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; (X.W.); (C.B.); (Z.L.)
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenjiang Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; (X.W.); (C.B.); (Z.L.)
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lupeng Yue
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; (X.W.); (C.B.); (Z.L.)
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: (L.Y.); (L.H.)
| | - Li Hu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; (X.W.); (C.B.); (Z.L.)
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: (L.Y.); (L.H.)
| |
Collapse
|
15
|
Sinha S, Tejus A, Mohan P, Mathur AG. Journey of oliceridine: A novel opioid. JOURNAL OF MARINE MEDICAL SOCIETY 2022. [DOI: 10.4103/jmms.jmms_174_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
16
|
Miller ER, Scheidt KA. Enantioselective Syntheses of Yohimbine Alkaloids: Proving Grounds for New Catalytic Transformations. SYNTHESIS-STUTTGART 2022; 54:1217-1230. [PMID: 36187077 PMCID: PMC9524398 DOI: 10.1055/a-1684-2942] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The total synthesis of bioactive alkaloids is an enduring challenge and an indication of the state of the art of chemical synthesis. With the explosion of catalytic asymmetric methods over the past three decades, these compelling targets have been fertile proving grounds for enantioselective bond forming transformations. These activities are summarized herein both to highlight the power and versatility of these methods and to instill future inspiration for new syntheses of these privileged natural products.
Collapse
|
17
|
Hernández-Alvarado RB, Madariaga-Mazón A, Cosme-Vela F, Marmolejo-Valencia AF, Nefzi A, Martinez-Mayorga K. Encoding mu-opioid receptor biased agonism with interaction fingerprints. J Comput Aided Mol Des 2021; 35:1081-1093. [PMID: 34713377 DOI: 10.1007/s10822-021-00422-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/07/2021] [Indexed: 10/20/2022]
Abstract
Opioids are potent painkillers, however, their therapeutic use requires close medical monitoring to diminish the risk of severe adverse effects. The G-protein biased agonists of the μ-opioid receptor (MOR) have shown safer therapeutic profiles than non-biased ligands. In this work, we performed extensive all-atom molecular dynamics simulations of two markedly biased ligands and a balanced reference molecule. From those simulations, we identified a protein-ligand interaction fingerprint that characterizes biased ligands. Then, we built and virtually screened a database containing 68,740 ligands with proven or potential GPCR agonistic activity. Exemplary molecules that fulfill the interacting pattern for biased agonism are showcased, illustrating the usefulness of this work for the search of biased MOR ligands and how this contributes to the understanding of MOR biased signaling.
Collapse
Affiliation(s)
| | | | - Fernando Cosme-Vela
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Adel Nefzi
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, USA
| | | |
Collapse
|
18
|
Muchhala KH, Jacob JC, Kang M, Dewey WL, Akbarali HI. The Guts of the Opioid Crisis. Physiology (Bethesda) 2021; 36:315-323. [PMID: 34431418 PMCID: PMC8813205 DOI: 10.1152/physiol.00014.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/10/2021] [Accepted: 06/10/2021] [Indexed: 01/09/2023] Open
Abstract
Bidirectional interactions of the gut epithelium with commensal bacteria are critical for maintaining homeostasis within the gut. Chronic opioid exposure perturbs gut homeostasis through a multitude of neuro-immune-epithelial mechanisms, resulting in the development of analgesic tolerance, a major underpinning of the current opioid crisis. Differences in molecular mechanisms of opioid tolerance between the enteric and central pain pathways pose a significant challenge for managing chronic pain without untoward gastrointestinal effects.
Collapse
Affiliation(s)
- Karan H Muchhala
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Joanna C Jacob
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Minho Kang
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - William L Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Hamid I Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
19
|
O'Brien JB, Roman DL. Novel treatments for chronic pain: moving beyond opioids. Transl Res 2021; 234:1-19. [PMID: 33727192 DOI: 10.1016/j.trsl.2021.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023]
Abstract
It is essential that safe and effective treatment options be available to patients suffering from chronic pain. The emergence of an opioid epidemic has shaped public opinions and created stigmas surrounding the use of opioids for the management of pain. This reality, coupled with high risk of adverse effects from chronic opioid use, has led chronic pain patients and their healthcare providers to utilize nonopioid treatment approaches. In this review, we will explore a number of cellular reorganizations that are associated with the development and progression of chronic pain. We will also discuss the safety and efficacy of opioid and nonopioid treatment options for chronic pain. Finally, we will review the evidence for adenylyl cyclase type 1 (AC1) as a novel target for the treatment of chronic pain.
Collapse
Affiliation(s)
- Joseph B O'Brien
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa
| | - David L Roman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa; Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
20
|
De Neve J, Barlow TMA, Tourwé D, Bihel F, Simonin F, Ballet S. Comprehensive overview of biased pharmacology at the opioid receptors: biased ligands and bias factors. RSC Med Chem 2021; 12:828-870. [PMID: 34223156 PMCID: PMC8221262 DOI: 10.1039/d1md00041a] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022] Open
Abstract
One of the main challenges in contemporary medicinal chemistry is the development of safer analgesics, used in the treatment of pain. Currently, moderate to severe pain is still treated with the "gold standard" opioids whose long-term often leads to severe side effects. With the discovery of biased agonism, the importance of this area of pharmacology has grown exponentially over the past decade. Of these side effects, tolerance, opioid misuse, physical dependence and substance use disorder (SUD) stand out, since these have led to many deaths over the past decades in both USA and Europe. New therapeutic molecules that induce a biased response at the opioid receptors (MOR, DOR, KOR and NOP receptor) are able to circumvent these side effects and, consequently, serve as more advantageous therapies with great promise. The concept of biased signaling extends far beyond the already sizeable field of GPCR pharmacology and covering everything would be vastly outside the scope of this review which consequently covers the biased ligands acting at the opioid family of receptors. The limitation of quantifying bias, however, makes this a controversial subject, where it is dependent on the reference ligand, the equation or the assay used for the quantification. Hence, the major issue in the field of biased ligands remains the translation of the in vitro profiles of biased signaling, with corresponding bias factors to in vivo profiles showing the presence or the lack of specific side effects. This review comprises a comprehensive overview of biased ligands in addition to their bias factors at individual members of the opioid family of receptors, as well as bifunctional ligands.
Collapse
Affiliation(s)
- Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Thomas M A Barlow
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Frédéric Bihel
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, CNRS Université de Strasbourg Illkirch France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242, CNRS, Université de Strasbourg Illkirch France
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| |
Collapse
|
21
|
Childers WE, Abou-Gharbia MA. "I'll Be Back": The Resurrection of Dezocine. ACS Med Chem Lett 2021; 12:961-968. [PMID: 34141081 DOI: 10.1021/acsmedchemlett.1c00233] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/07/2021] [Indexed: 12/25/2022] Open
Abstract
Beginning with opium itself, natural and synthetic opioids have been used as analgesics for over 8000 years and were likely abused as drugs of recreation for that long as well. However, the "opioid crisis" resulted in attempts to avoid or limit opioid analgesics in favor of other therapies and methods. Mu opioid agonists can be effective analgesics but suffer from addiction, tolerance, and dangerous, sometimes fatal, side effects. One exception to this generalization is dezocine (Dalgan), a mixed mu/kappa opioid partial agonist. Dezocine is at least as effective as morphine in reducing acute pain in animal models and clinical applications such as postoperative pain. And while dezocine was discontinued in western markets in 2011, it has become the favored opioid analgesic in China, capturing over 40% of the market. Additionally, dezocine possesses norepinephrine uptake inhibitory activity, which may synergize with mu agonism in the case of acute pain treatment and possibly endow the drug with antinociceptive activity in neuropathic pain conditions. This Innovations article summarizes the history and properties of dezocine and presents evidence and rationale for why dezocine has undergone a resurrection.
Collapse
Affiliation(s)
- Wayne E. Childers
- The Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, Pennsylvania 19140, United States
| | - Magid A. Abou-Gharbia
- The Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
22
|
A Large-Scale Observational Study on the Temporal Trends and Risk Factors of Opioid Overdose: Real-World Evidence for Better Opioids. Drugs Real World Outcomes 2021; 8:393-406. [PMID: 34037960 PMCID: PMC8324607 DOI: 10.1007/s40801-021-00253-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 11/25/2022] Open
Abstract
Background The USA is in the midst of an opioid overdose epidemic. To address the epidemic, we conducted a large-scale population study on opioid overdose. Objectives The primary objective of this study was to evaluate the temporal trends and risk factors of inpatient opioid overdose. Based on its patterns, the secondary objective was to examine the innate properties of opioid analgesics underlying reduced overdose effects. Methods A retrospective cross-sectional study was conducted based on a large-scale inpatient electronic health records database, Cerner Health Facts®, with (1) inclusion criteria for participants as patients admitted between 1 January, 2009 and 31 December, 2017 and (2) measurements as opioid overdose prevalence by year, demographics, and prescription opioid exposures. Results A total of 4,720,041 patients with 7,339,480 inpatient encounters were retrieved from Cerner Health Facts®. Among them, 30.2% patients were aged 65+ years, 57.0% female, 70.1% Caucasian, 42.3% single, 32.0% from the South, and 80.8% in an urban area. From 2009 to 2017, annual opioid overdose prevalence per 1000 patients significantly increased from 3.7 to 11.9 with an adjusted odds ratio (aOR): 1.16, 95% confidence interval (CI) 1.15–1.16. Compared to the major demographic counterparts, being in (1) age group: 41–50 years (overall aOR 1.36, 95% CI 1.31–1.40) or 51–64 years (overall aOR 1.35, 95% CI 1.32–1.39), (2) marital status: divorced (overall aOR 1.19, 95% CI 1.15–1.23), and (3) census region: West (overall aOR 1.32, 95% CI 1.28–1.36) were significantly associated with a higher odds of opioid overdose. Prescription opioid exposures were also associated with an increased odds of opioid overdose, such as meperidine (overall aOR 1.09, 95% CI 1.06–1.13) and tramadol (overall aOR 2.20, 95% CI 2.14–2.27). Examination on the relationships between opioid analgesic properties and their association strengths, aORs, and opioid overdose showed that lower aOR values were significantly associated with (1) high molecular weight, (2) non-interaction with multi-drug resistance protein 1 or interaction with cytochrome P450 3A4, and (3) non-interaction with the delta opioid receptor or kappa opioid receptor. Conclusions The significant increasing trends of opioid overdose at the inpatient care setting from 2009 to 2017 suggested an ongoing need for efforts to combat the opioid overdose epidemic in the USA. Risk factors associated with opioid overdose included patient demographics and prescription opioid exposures. Moreover, there are physicochemical, pharmacokinetic, and pharmacodynamic properties underlying reduced overdose effects, which can be utilized to develop better opioids. Supplementary Information The online version contains supplementary material available at 10.1007/s40801-021-00253-8.
Collapse
|
23
|
Muchhala KH, Jacob JC, Dewey WL, Akbarali HI. Role of β-arrestin-2 in short- and long-term opioid tolerance in the dorsal root ganglia. Eur J Pharmacol 2021; 899:174007. [PMID: 33705801 PMCID: PMC8058323 DOI: 10.1016/j.ejphar.2021.174007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/25/2021] [Accepted: 02/28/2021] [Indexed: 11/26/2022]
Abstract
G-protein-biased agonists with reduced β-arrestin-2 activation are being investigated as safer alternatives to clinically-used opioids. β-arrestin-2 has been implicated in the mechanism of opioid-induced antinociceptive tolerance. Opioid-induced analgesic tolerance is classically considered as centrally-mediated, but recent reports implicate nociceptive dorsal root ganglia neurons as critical mediators in this process. Here, we investigated the role of β-arrestin-2 in the mechanism of opioid tolerance in dorsal root ganglia nociceptive neurons using β-arrestin-2 knockout mice and the G-protein-biased μ-opioid receptor agonist, TRV130. Whole-cell current-clamp electrophysiology experiments revealed that 15-18-h overnight exposure to 10 μM morphine in vitro induced acute tolerance in β-arrestin-2 wild-type but not knockout neurons. Furthermore, in wild-type neurons circumventing β-arrestin-2 activation by overnight treatment with 200 nM TRV130 attenuated tolerance. Similarly, acute morphine tolerance in vivo in β-arrestin-2 knockout mice was prevented in the warm-water tail-withdrawal assay. Treatment with 30 mg/kg TRV130 s.c. also inhibited acute antinociceptive tolerance in vivo in wild-type mice. Alternately, in β-arrestin-2 knockout neurons tolerance induced by 7-day in vivo exposure to 50 mg morphine pellet was conserved. Likewise, β-arrestin-2 deletion did not mitigate in vivo antinociceptive tolerance induced by 7-day exposure to 25 mg or 50 mg morphine pellet in both female or male mice, respectively. Consequently, these results indicated that β-arrestin-2 mediates acute but not chronic opioid tolerance in dorsal root ganglia neurons and to antinociception in vivo. This suggests that opioid-induced antinociceptive tolerance may develop even in the absence of β-arrestin-2 activation, and thus significantly affect the clinical utility of biased agonists.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Behavior, Animal/drug effects
- Cells, Cultured
- Disease Models, Animal
- Drug Tolerance
- Female
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/physiopathology
- Male
- Mice, Knockout
- Morphine/pharmacology
- Neurons/drug effects
- Neurons/metabolism
- Nociceptive Pain/genetics
- Nociceptive Pain/metabolism
- Nociceptive Pain/physiopathology
- Nociceptive Pain/prevention & control
- Pain Threshold/drug effects
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Spiro Compounds/pharmacology
- Thiophenes/pharmacology
- Time Factors
- beta-Arrestin 2/deficiency
- beta-Arrestin 2/genetics
- beta-Arrestin 2/metabolism
- Mice
Collapse
Affiliation(s)
- Karan H Muchhala
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1112 East Clay St, Richmond, VA, 23298, USA
| | - Joanna C Jacob
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1112 East Clay St, Richmond, VA, 23298, USA
| | - William L Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1112 East Clay St, Richmond, VA, 23298, USA
| | - Hamid I Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1112 East Clay St, Richmond, VA, 23298, USA.
| |
Collapse
|
24
|
Zhang JJ, Song CG, Dai JM, Zhang XQ, Lin P, Li L, Yang XM, Chen ZN. Inhibition of mu-opioid receptor suppresses proliferation of hepatocellular carcinoma cells via CD147-p53-MAPK cascade signaling pathway. Am J Transl Res 2021; 13:3967-3986. [PMID: 34149993 PMCID: PMC8205673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/18/2021] [Indexed: 06/12/2023]
Abstract
Hepatocellular carcinoma (HCC) is the leading cause of cancer-related deaths. Previous studies have suggested that mu-opioid receptor (MOR), a member of the opioid receptor family, is involved in the pathogenesis of HCC. However, the mechanism by which MOR regulates the biological behavior of HCC is still poorly understood. To address this problem, in this study, we investigated the role of MOR in the proliferation of HCC cell lines and the underlying mechanism. First, RT-PCR, western-blot and immunohistochemistry revealed higher expression of MOR in HCC cells and tissue than in non-tumor cells or adjacent tissue, and elevated expression of MOR was associated with jeopardized survival of HCC patients, as demonstrated by bioinformatic databases. Knockdown of MOR by specific siRNA attenuated the proliferation and migration of HCC cells and this effect could be reversed by rescue experiments, confirming the essential role of MOR in the proliferation of HCC. Moreover, results of colony formation assay, CCK8 test, flow cytometry and western blot suggested that a monoclonal antibody (mAb) specifically against MOR could inhibit proliferation of HepG2 and Huh7 cells via the MOR-CD147-p53-MAPK pathway, and the interaction between MOR and CD147 was verified by immunofluorescence colocalization and co-IP analysis. The mAb against MOR also enhanced the cisplatin-induced apoptosis of HCC cells by downregulating p-ERK, Bcl-2 and upregulating Bax. Taken together, these results suggest that MOR could regulate the proliferation of HCC cells in a CD147-p53-MAPK dependent manner. MOR possesses the potential to be a therapeutic target to treat HCC.
Collapse
Affiliation(s)
- Jia-Jia Zhang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, The Fourth Military Medical UniversityXi’an 710032, China
| | - Chang-Geng Song
- Department of Neurology, Xijing Hospital, The Fourth Military Medical UniversityXi’an 710032, China
| | - Ji-Min Dai
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical UniversityXi’an 710032, China
| | - Xue-Qin Zhang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, The Fourth Military Medical UniversityXi’an 710032, China
| | - Peng Lin
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, The Fourth Military Medical UniversityXi’an 710032, China
| | - Ling Li
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, The Fourth Military Medical UniversityXi’an 710032, China
| | - Xiang-Min Yang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, The Fourth Military Medical UniversityXi’an 710032, China
| | - Zhi-Nan Chen
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, The Fourth Military Medical UniversityXi’an 710032, China
| |
Collapse
|
25
|
Manning JJ, Green HM, Glass M, Finlay DB. Pharmacological selection of cannabinoid receptor effectors: Signalling, allosteric modulation and bias. Neuropharmacology 2021; 193:108611. [PMID: 34000272 DOI: 10.1016/j.neuropharm.2021.108611] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/26/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022]
Abstract
The type-1 cannabinoid receptor (CB1) is a promising drug target for a wide range of diseases. However, many existing and novel candidate ligands for CB1 have shown only limited therapeutic potential. Indeed, no ligands are currently approved for the clinic except formulations of the phytocannabinoids Δ9-THC and CBD and a small number of analogues. A key limitation of many promising CB1 ligands are their on-target adverse effects, notably including psychoactivity (agonists) and depression/suicidal ideation (inverse agonists). Recent drug development attempts have therefore focussed on altering CB1 signalling profiles in two ways. Firstly, with compounds that enhance or reduce the signalling of endogenous (endo-) cannabinoids, namely allosteric modulators. Secondly, with compounds that probe the capability of selectively targeting specific cellular signalling pathways that may mediate therapeutic effects using biased ligands. This review will summarise the current paradigm of CB1 signalling in terms of the intracellular transduction pathways acted on by the receptor. The development of compounds that selectively activate CB1 signalling pathways, whether allosterically or via orthosteric agonist bias, will also be addressed.
Collapse
Affiliation(s)
- Jamie J Manning
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand, PO Box 56, Dunedin, 9054, New Zealand
| | - Hayley M Green
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand, PO Box 56, Dunedin, 9054, New Zealand
| | - Michelle Glass
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand, PO Box 56, Dunedin, 9054, New Zealand
| | - David B Finlay
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand, PO Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
26
|
Webster L, Schmidt WK. Dilemma of Addiction and Respiratory Depression in the Treatment of Pain: A Prototypical Endomorphin as a New Approach. PAIN MEDICINE 2021; 21:992-1004. [PMID: 31165885 DOI: 10.1093/pm/pnz122] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Although mu-opioid receptor agonists have been the mainstay of analgesic regimens for moderate to severe pain, they are associated with serious side effects, risks, and limitations. We evaluate the most serious risks associated with conventional opioids and compare these with the pharmacology of CYT-1010, a prototypical endomorphin and mu-opioid receptor agonist. RESULTS Addiction and respiratory depression are serious risks of traditional mu-opioid analgesics. Mitigation strategies have been inadequate at addressing the opioid crisis and may interfere with the effective treatment of pain. Improved understanding of mu-opioid receptor biology and the discovery in 1997 of an additional and unique family of endogenous opioid peptides (endomorphins) have provided a pathway for dissociating analgesia from opioid-related adverse events and developing new classes of mu-opioid receptor agonists that use biased signaling and/or target novel sites to produce analgesia with reduced side effect liability. Endomorphin-1 and -2 are endogenous opioid peptides highly selective for mu-opioid receptors that exhibit potent analgesia with reduced side effects. CYT-1010 is a cyclized, D-lysine-containing analog of endomorphin-1 with a novel mechanism of action targeting traditional mu- and exon 11/truncated mu-opioid receptor 6TM variants. CYT-1010 preclinical data have demonstrated reduced abuse potential and analgesic potency exceeding that of morphine. In an initial phase 1 clinical study, CYT-1010 demonstrated significant analgesia vs baseline and no respiratory depression at the dose levels tested. CONCLUSIONS CYT-1010 and other novel mu-opioid receptor agonists in clinical development are promising alternatives to conventional opioids that may offer the possibility of safer treatment of moderate to severe pain.
Collapse
|
27
|
Capsazepine decreases corneal pain syndrome in severe dry eye disease. J Neuroinflammation 2021; 18:111. [PMID: 33975636 PMCID: PMC8114509 DOI: 10.1186/s12974-021-02162-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/29/2021] [Indexed: 11/26/2022] Open
Abstract
Background Dry eye disease (DED) is a multifactorial disease of the ocular surface accompanied by neurosensory abnormalities. Here, we evaluated the effectiveness of transient receptor potential vanilloid-1 (TRPV1) blockade to alleviate ocular pain, neuroinflammation, and anxiety-like behavior associated with severe DED. Methods Chronic DED was induced by unilateral excision of the Harderian and extraorbital lacrimal glands of adult male mice. Investigations were conducted at 21 days after surgery. The mRNA levels of TRPV1, transient receptor potential ankyrin-1 (TRPA1), and acid-sensing ion channels 1 and 3 (ASIC1 and ASIC3) in the trigeminal ganglion (TG) were evaluated by RNAscope in situ hybridization. Multi-unit extracellular recording of ciliary nerve fiber activity was used to monitor spontaneous and stimulated (cold, heat, and acid) corneal nerve responsiveness in ex vivo eye preparations. DED mice received topical instillations of the TRPV1 antagonist (capsazepine) twice a day for 2 weeks from d7 to d21 after surgery. The expression of genes involved in neuropathic and inflammatory pain was evaluated in the TG using a global genomic approach. Chemical and mechanical corneal nociception and spontaneous ocular pain were monitored. Finally, anxiety-like behaviors were assessed by elevated plus maze and black and white box tests. Results First, in situ hybridization showed DED to trigger upregulation of TRPV1, TRPA1, ASIC1, and ASIC3 mRNA in the ophthalmic branch of the TG. DED also induced overexpression of genes involved in neuropathic and inflammatory pain in the TG. Repeated instillations of capsazepine reduced corneal polymodal responsiveness to heat, cold, and acidic stimulation in ex vivo eye preparations. Consistent with these findings, chronic capsazepine instillation inhibited the upregulation of genes involved in neuropathic and inflammatory pain in the TG of DED animals and reduced the sensation of ocular pain, as well as anxiety-like behaviors associated with severe DED. Conclusion These data provide novel insights on the effectiveness of TRPV1 antagonist instillation in alleviating abnormal corneal neurosensory symptoms induced by severe DED, opening an avenue for the repositioning of this molecule as a potential analgesic treatment for patients suffering from chronic DED.
Collapse
|
28
|
Marmolejo-Valencia AF, Madariaga-Mazón A, Martinez-Mayorga K. Bias-inducing allosteric binding site in mu-opioid receptor signaling. SN APPLIED SCIENCES 2021. [DOI: 10.1007/s42452-021-04505-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Abstract
G-protein-biased agonism of the mu-opioid receptor (μ-OR) is emerging as a promising strategy in analgesia. A deep understanding of how biased agonists modulate and differentiate G-protein-coupled receptors (GPCR) signaling pathways and how this is transferred into the cell are open questions. Here, using extensive all-atom molecular dynamics simulations, we analyzed the binding recognition process and signaling effects of three prototype μ-OR agonists. Our suggested structural mechanism of biased signaling in μ-OR involves an allosteric sodium ion site, water networks, conformational rearrangements in conserved motifs and collective motions of loops and transmembrane helices. These analyses led us to highlight the relevance of a bias-inducing allosteric binding site in the understanding of μ-OR’s G-protein-biased signaling. These results also suggest a competitive equilibrium between the agonists and the allosteric sodium ion, where the bias-inducing allosteric binding site can be modulated by this ion or an agonist such as herkinorin. Notably, herkinorin arises as the archetype modulator of μ-OR and its interactive pattern could be used for screening efforts via protein–ligand interaction fingerprint (PLIF) studies.
Article highlights
Agonists and a sodium ion compete for the bias-inducing allosteric binding site that modulates signaling in mu-opioid receptors.
Molecular dynamics simulations of the prototype μ-OR agonist suggest a competitive equilibrium involving the agonist and an allosteric sodium ion.
Analysis of experimental data from the literature and molecular models provides the structural bases of biased agonism on μ-OR.
Collapse
|
29
|
Functional approaches to the study of G-protein-coupled receptors in postmortem brain tissue: [ 35S]GTPγS binding assays combined with immunoprecipitation. Pharmacol Rep 2021; 73:1079-1095. [PMID: 33876404 DOI: 10.1007/s43440-021-00253-z] [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: 02/08/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 10/21/2022]
Abstract
G-protein-coupled receptors (GPCRs) have an enormous biochemical importance as they bind to diverse extracellular ligands and regulate a variety of physiological and pathological responses. G-protein activation measures the functional consequence of receptor occupancy at one of the earliest receptor-mediated events. Receptor coupling to G-proteins promotes the GDP/GTP exchange on Gα subunits. Thus, modulation of the binding of the poorly hydrolysable GTP analog [35S]GTPγS to the Gα-protein subunit can be used as a functional approach to quantify GPCR interaction with agonist, antagonist or inverse agonist drugs. In order to determine receptor-mediated selective activation of the different Gα-proteins, [35S]GTPγS binding assays combined with immunodetection by specific antibodies have been developed and applied to physiological and pathological brain conditions. Currently, immunoprecipitation with magnetic beads and scintillation proximity assays are the most habitual techniques for this purpose. The present review summarizes the different procedures, advantages and limitations of the [35S]GTPγS binding assays combined with selective Gα-protein sequestration methods. Experience of functional coupling of several GPCRs to different Gα-proteins and recommendations for optimal performance in brain membranes are described. One of the biggest opportunities opened by these techniques is that they enable evaluation of biased agonism in the native tissue, which results in high interest in drug discovery. The available results derived from application of these functional methodologies to study GPCR dysfunctions in neuro-psychiatric disorders are also described. In conclusion, [35S]GTPγS binding combined with antibody-mediated immunodetection represents an useful method to separately evaluate the functional activity of drugs acting on GPCRs over each Gα-protein subtype.
Collapse
|
30
|
Aghajani N, Pourhamzeh M, Azizi H, Semnanian S. Central blockade of orexin type 1 receptors reduces naloxone induced activation of locus coeruleus neurons in morphine dependent rats. Neurosci Lett 2021; 755:135909. [PMID: 33892002 DOI: 10.1016/j.neulet.2021.135909] [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: 02/19/2021] [Revised: 04/04/2021] [Accepted: 04/16/2021] [Indexed: 11/30/2022]
Abstract
Orexin neuropeptides are implicated in the expression of morphine dependence. Locus coeruleus (LC) nucleus is an important brain area involving in the development of withdrawal signs of morphine and contains high expression of orexin type 1 receptors (OX1Rs). Despite extensive considerations, effects of immediate inhibition of OX1Rs by a single dose administration of SB-334867 prior to the naloxone-induced activation of LC neurons remains unknown. Therefore, we examined the direct effects of OX1Rs acute blockade on the neuronal activity of the morphine-dependent rats which underwent naloxone administration. Adult male rats underwent subcutaneous administration of 10 mg/kg morphine (two times/day) for a ten-day period. On the last day of experiment, intra-cerebroventricular administration of 10 μg/μl antagonist of OX1Rs, SB-334867, was performed just before intra-peritoneal injection of 2 mg/kg naloxone. Thereafter, in vivo extracellular single unit recording was employed to evaluate the electrical activity of LC neuronal cells. The outcomes demonstrated that morphine tolerance developed following ten-day of injection. Then, naloxone administration causes hyperactivity of LC neuronal cells, whereas a single dose administration of SB-334867 prior to naloxone prevented the enhanced activity of neurons upon morphine withdrawal. Our findings indicate that increased response of LC neuronal cells to applied naloxone could be prevented by the acute inhibition of the OX1Rs just before the naloxone treatment.
Collapse
Affiliation(s)
- Niloofar Aghajani
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahsa Pourhamzeh
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
31
|
Jia X, Ciallella HL, Russo DP, Zhao L, James MH, Zhu H. Construction of a Virtual Opioid Bioprofile: A Data-Driven QSAR Modeling Study to Identify New Analgesic Opioids. ACS SUSTAINABLE CHEMISTRY & ENGINEERING 2021; 9:3909-3919. [PMID: 34239782 PMCID: PMC8259887 DOI: 10.1021/acssuschemeng.0c09139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Compared to traditional experimental approaches, computational modeling is a promising strategy to efficiently prioritize new candidates with low cost. In this study, we developed a novel data mining and computational modeling workflow proven to be applicable by screening new analgesic opioids. To this end, a large opioid data set was used as the probe to automatically obtain bioassay data from the PubChem portal. There were 114 PubChem bioassays selected to build quantitative structure-activity relationship (QSAR) models based on the testing results across the probe compounds. The compounds tested in each bioassay were used to develop 12 models using the combination of three machine learning approaches and four types of chemical descriptors. The model performance was evaluated by the coefficient of determination (R 2) obtained from 5-fold cross-validation. In total, 49 models developed for 14 bioassays were selected based on the criteria and were identified to be mainly associated with binding affinities to different opioid receptors. The models for these 14 bioassays were further used to fill data gaps in the probe opioids data set and to predict general drug compounds in the DrugBank data set. This study provides a universal modeling strategy that can take advantage of large public data sets for computer-aided drug design (CADD).
Collapse
Affiliation(s)
- Xuelian Jia
- The Rutgers Center for Computational and Integrative Biology, Joint Health Sciences Center, Camden, New Jersey 08103, United States
| | - Heather L Ciallella
- The Rutgers Center for Computational and Integrative Biology, Joint Health Sciences Center, Camden, New Jersey 08103, United States
| | - Daniel P Russo
- The Rutgers Center for Computational and Integrative Biology, Joint Health Sciences Center, Camden, New Jersey 08103, United States
| | - Linlin Zhao
- The Rutgers Center for Computational and Integrative Biology, Joint Health Sciences Center, Camden, New Jersey 08103, United States
| | - Morgan H James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, New Jersey 08854, United States; Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, New Jersey 08854, United States
| | - Hao Zhu
- The Rutgers Center for Computational and Integrative Biology, Joint Health Sciences Center, Camden, New Jersey 08103, United States; Department of Chemistry, Rutgers University, Camden, New Jersey 08102, United States
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Opioid use disorder (OUD) remains a national epidemic with an immense consequence to the United States' healthcare system. Current therapeutic options are limited by adverse effects and limited efficacy. RECENT FINDINGS Recent advances in therapeutic options for OUD have shown promise in the fight against this ongoing health crisis. Modifications to approved medication-assisted treatment (MAT) include office-based methadone maintenance, implantable and monthly injectable buprenorphine, and an extended-release injectable naltrexone. Therapies under investigation include various strategies such as heroin vaccines, gene-targeted therapy, and biased agonism at the G protein-coupled receptor (GPCR), but several pharmacologic, clinical, and practical barriers limit these treatments' market viability. This manuscript provides a comprehensive review of the current literature regarding recent innovations in OUD treatment.
Collapse
|
33
|
Ehrlich AT, Darcq E. Recent advances in basic science methodology to evaluate opioid safety profiles and to understand opioid activities. Fac Rev 2021; 10:15. [PMID: 33718932 PMCID: PMC7946392 DOI: 10.12703/r/10-15] [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] [Indexed: 12/02/2022] Open
Abstract
Opioids are powerful drugs used by humans for centuries to relieve pain and are still frequently used as pain treatment in current clinical practice. Medicinal opioids primarily target the mu opioid receptor (MOR), and MOR activation produces unmatched pain-alleviating properties, as well as side effects such as strong rewarding effects, and thus abuse potential, and respiratory depression contributing to death during overdose. Therefore, the ultimate goal is to create opioid pain-relievers with reduced respiratory depression and thus fewer chances of lethality. Efforts are also underway to reduce the euphoric effects of opioids and avoid abuse liability. In this review, recent advances in basic science methodology used to understand MOR pharmacology and activities will be summarized. The focus of the review will be to describe current technological advances that enable the study of opioid analgesics from subcellular mechanisms to mesoscale network responses. These advances in understanding MOR physiological responses will help to improve knowledge and future design of opioid analgesics.
Collapse
Affiliation(s)
- Aliza T Ehrlich
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Emmanuel Darcq
- Department of Psychiatry, Douglas Research Center, McGill University, Montréal, Canada
- INSERM U1114, UNISTRA University of Strasbourg, Strasbourg, France
| |
Collapse
|
34
|
Liao S, Tan K, Floyd C, Bong D, Pino MJ, Wu C. Probing biased activation of mu-opioid receptor by the biased agonist PZM21 using all atom molecular dynamics simulation. Life Sci 2021; 269:119026. [PMID: 33444617 DOI: 10.1016/j.lfs.2021.119026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/25/2020] [Accepted: 01/02/2021] [Indexed: 01/14/2023]
Abstract
Morphine is a commonly used opioid drug to treat acute pain by binding to the mu-opioid receptor (MOR), but its effective analgesic efficacy via triggering of the heterotrimeric Gi protein pathway is accompanied by a series of adverse side effects via triggering of the β-arrestin pathway. Recently, PZM21, a recently developed MOR biased agonist, shows preferentially activating the G protein pathway over β-arrestin pathway. However, there is no high-resolution receptor structure in complex with PZM21 and its action mechanism remains elusive. In this study, PZM21 and Morphine were docked to the active human MOR-1 homology structure and then subjected to the molecular dynamics (MD) simulations in two different situations (i.e., one situation includes the crystal waters but another does not). Detailed comparisons between the two systems were made to characterize the differences in protein-ligand interactions, protein secondary and tertiary structures and dynamics networks. PZM21 could strongly interact with Y3287.43 of TM7, besides the residues (Asp1493.32 and Tyr1503.33) of TM3. The two systems' network paths to the intracellular end of TM6 were roughly similar but the paths to the end of TM7 were different. The PZM21-bound MOR's intracellular ends of TM5-7 bent outward more along with the distance changes of the three key molecular switches (ionic lock, transmission and Tyr toggle) and the distance increase of some conserved inter-helical residue pairs. The larger intracellular opening of the receptor could potentially facilitate G protein binding.
Collapse
Affiliation(s)
- Siyan Liao
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China; College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Kai Tan
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA; Department of Chemistry, Xiamen University, Xiamen, Fujian 361005, China
| | - Cecilia Floyd
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Daegun Bong
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Michael James Pino
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Chun Wu
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA.
| |
Collapse
|
35
|
Pineyro G, Nagi K. Signaling diversity of mu- and delta- opioid receptor ligands: Re-evaluating the benefits of β-arrestin/G protein signaling bias. Cell Signal 2020; 80:109906. [PMID: 33383156 DOI: 10.1016/j.cellsig.2020.109906] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/24/2020] [Accepted: 12/27/2020] [Indexed: 01/02/2023]
Abstract
Opioid analgesics are elective for treating moderate to severe pain but their use is restricted by severe side effects. Signaling bias has been proposed as a viable means for improving this situation. To exploit this opportunity, continuous efforts are devoted to understand how ligand-specific modulations of receptor functions could mediate the different in vivo effects of opioids. Advances in the field have led to the development of biased agonists based on hypotheses that allocated desired and undesired effects to specific signaling pathways. However, the prevalent hypothesis associating β-arrestin to opioid side effects was recently challenged and multiple of the newly developed biased drugs may not display the superior side effects profile that was sought. Moreover, biased agonism at opioid receptors is now known to be time- and cell-dependent, which adds a new layer of complexity for bias estimation. Here, we first review the signaling mechanisms underlying desired and undesired effects of opioids. We then describe biased agonism at opioid receptors and discuss the different perspectives that support the desired and undesired effects of opioids in view of exploiting biased signaling for therapeutic purposes. Finally, we explore how signaling kinetics and cellular background can influence the magnitude and directionality of bias at those receptors.
Collapse
Affiliation(s)
- Graciela Pineyro
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; CHU Sainte-Justine research center, Montreal, QC H3T 1C5, Canada
| | - Karim Nagi
- College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
36
|
Hernández-Alvarado RB, Madariaga-Mazón A, Ortega A, Martinez-Mayorga K. DARK Classics in Chemical Neuroscience: Salvinorin A. ACS Chem Neurosci 2020; 11:3979-3992. [PMID: 33164503 DOI: 10.1021/acschemneuro.0c00608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Salvinorin A is the main bioactive compound in Salvia divinorum, an endemic plant with ancestral use by the inhabitants of the Mazateca mountain range (Sierra Mazateca) in Oaxaca, México. The main use of la pastora, as locally known, is in spiritual rites due to its extraordinary hallucinogenic effects. Being the first known nonalkaloidal opioid-mediated psychotropic molecule, salvinorin A set new research areas in neuroscience. The absence of a protonated amine group, common to all previously known opioids, results in a fast metabolism with the concomitant fast elimination and swift loss of activity. The worldwide spread and psychotropic effects of salvinorin A account for its misuse and classification as a drug of abuse. Consequently, salvinorin A and Salvia divinorum are now banned in many countries. Several synthetic efforts have been focused on the improvement of physicochemical and biological properties of salvinorin A: from total synthesis to hundreds of analogues. In this Review, we discuss the impact of salvinorin A in chemistry and neuroscience covering the historical relevance, isolation from natural sources, synthetic efforts, and pharmacological and safety profiles. Altogether, the chemistry behind and the taboo that encloses salvinorin A makes it one of the most exquisite naturally occurring drugs.
Collapse
Affiliation(s)
- R. Bruno Hernández-Alvarado
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México, 04510 México
| | - Abraham Madariaga-Mazón
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México, 04510 México
| | - Alfredo Ortega
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México, 04510 México
| | - Karina Martinez-Mayorga
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México, 04510 México
| |
Collapse
|
37
|
Maxwell EA, King TI, Kamble SH, Raju KSR, Berthold EC, León F, Avery BA, McMahon LR, McCurdy CR, Sharma A. Pharmacokinetics and Safety of Mitragynine in Beagle Dogs. PLANTA MEDICA 2020; 86:1278-1285. [PMID: 32693425 PMCID: PMC7907416 DOI: 10.1055/a-1212-5475] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Mitragynine is the most abundant psychoactive alkaloid derived from the leaves of Mitragyna speciosa (kratom), a tropical plant indigenous to regions of Southeast Asia. Mitragynine displays a moderate affinity to opioid receptors, and kratom is often self-prescribed to treat pain and/or opioid addiction. The purpose of this study was to investigate the safety and pharmacokinetic properties of mitragynine in the dog. Single dose oral (5 mg/kg) and intravenous (0.1 mg/kg) pharmacokinetic studies of mitragynine were performed in female beagle dogs. The plasma concentrations of mitragynine were measured using ultra-performance liquid chromatography coupled with a tandem mass spectrometer, and the pharmacokinetic properties were analyzed using non-compartmental analysis. Following intravenous administration, mitragynine showed a large volume of distribution (Vd, 6.3 ± 0.6 L/kg) and high clearance (Cl, 1.8 ± 0.4 L/h/kg). Following oral mitragynine dosing, first peak plasma (Cmax, 278.0 ± 47.4 ng/mL) concentrations were observed within 0.5 h. A potent mu-opioid receptor agonist and active metabolite of mitragynine, 7-hydroxymitragynine, was also observed with a Cmax of 31.5 ± 3.3 ng/mL and a Tmax of 1.7 ± 0.6 h in orally dosed dogs while its plasma concentrations were below the lower limit of quantification (1 ng/mL) for the intravenous study. The absolute oral bioavailability of mitragynine was 69.6%. Administration of mitragynine was well tolerated, although mild sedation and anxiolytic effects were observed. These results provide the first detailed pharmacokinetic information for mitragynine in a non-rodent species (the dog) and therefore also provide significant information for allometric scaling and dose predictions when designing clinical studies.
Collapse
Affiliation(s)
- Elizabeth A. Maxwell
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Tamara I. King
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Shyam H. Kamble
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Translational Drug Development Core, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, USA
| | - Kanumuri Siva Rama Raju
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Translational Drug Development Core, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, USA
| | - Erin C. Berthold
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Francisco León
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Bonnie A. Avery
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Translational Drug Development Core, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, USA
| | - Lance R. McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Christopher R. McCurdy
- Translational Drug Development Core, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, USA
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Abhisheak Sharma
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Translational Drug Development Core, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
38
|
Miyano K, Manabe S, Komatsu A, Fujii Y, Mizobuchi Y, Uezono E, Ohshima K, Nonaka M, Kuroda Y, Narita M, Uezono Y. The G Protein Signal-Biased Compound TRV130; Structures, Its Site of Action and Clinical Studies. Curr Top Med Chem 2020; 20:2822-2829. [PMID: 33115393 DOI: 10.2174/1568026620999201027224229] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 11/22/2022]
Abstract
Opioid agonists elicit their analgesic action mainly via μ opioid receptors; however, their use is limited because of adverse events including constipation and respiratory depression. It has been shown that analgesic action is transduced by the G protein-mediated pathway whereas adverse events are by the β-arrestin-mediated pathway through μ opioid receptor signaling. The first new-generation opioid TRV130, which preferentially activates G protein- but not β-arrestin-mediated signal, was constructed and developed to reduce adverse events. TRV130 and other G protein-biased compounds tend to elicit desirable analgesic action with less adverse effects. In clinical trials, the intravenous TRV130 (oliceridine) was evaluated in Phase I, II and III clinical studies. Here we review the discovery and synthesis of TRV130, its main action as a novel analgesic having less adverse events, its up-to-date status in clinical trials, and additional concerns about TRV130 as demonstrated in the literature.
Collapse
Affiliation(s)
- Kanako Miyano
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Sei Manabe
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Akane Komatsu
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuriko Fujii
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yusuke Mizobuchi
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Eiko Uezono
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kaori Ohshima
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Miki Nonaka
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yui Kuroda
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo,, Japan
| | - Minoru Narita
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasuhito Uezono
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
39
|
Thibeault PE, Ramachandran R. Biased signaling in platelet G-protein coupled receptors. Can J Physiol Pharmacol 2020; 99:255-269. [PMID: 32846106 DOI: 10.1139/cjpp-2020-0149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Platelets are small megakaryocyte-derived, anucleate, disk-like structures that play an outsized role in human health and disease. Both a decrease in the number of platelets and a variety of platelet function disorders result in petechiae or bleeding that can be life threatening. Conversely, the inappropriate activation of platelets, within diseased blood vessels, remains the leading cause of death and morbidity by affecting heart attacks and stroke. The fine balance of the platelet state in healthy individuals is controlled by a number of receptor-mediated signaling pathways that allow the platelet to rapidly respond and maintain haemostasis. G-protein coupled receptors (GPCRs) are particularly important regulators of platelet function. Here we focus on the major platelet-expressed GPCRs and discuss the roles of downstream signaling pathways (e.g., different G-protein subtypes or β-arrestin) in regulating the different phases of the platelet activation. Further, we consider the potential for selectively targeting signaling pathways that may contribute to platelet responses in disease through development of biased agonists. Such selective targeting of GPCR-mediated signaling pathways by drugs, often referred to as biased signaling, holds promise in delivering therapeutic interventions that do not present significant side effects, especially in finely balanced physiological systems such as platelet activation in haemostasis.
Collapse
Affiliation(s)
- Pierre E Thibeault
- Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street, London, ON N6A5C1, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street, London, ON N6A5C1, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street, London, ON N6A5C1, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street, London, ON N6A5C1, Canada
| |
Collapse
|
40
|
Azevedo Neto J, Costanzini A, De Giorgio R, Lambert DG, Ruzza C, Calò G. Biased versus Partial Agonism in the Search for Safer Opioid Analgesics. Molecules 2020; 25:molecules25173870. [PMID: 32854452 PMCID: PMC7504468 DOI: 10.3390/molecules25173870] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 02/07/2023] Open
Abstract
Opioids such as morphine—acting at the mu opioid receptor—are the mainstay for treatment of moderate to severe pain and have good efficacy in these indications. However, these drugs produce a plethora of unwanted adverse effects including respiratory depression, constipation, immune suppression and with prolonged treatment, tolerance, dependence and abuse liability. Studies in β-arrestin 2 gene knockout (βarr2(−/−)) animals indicate that morphine analgesia is potentiated while side effects are reduced, suggesting that drugs biased away from arrestin may manifest with a reduced-side-effect profile. However, there is controversy in this area with improvement of morphine-induced constipation and reduced respiratory effects in βarr2(−/−) mice. Moreover, studies performed with mice genetically engineered with G-protein-biased mu receptors suggested increased sensitivity of these animals to both analgesic actions and side effects of opioid drugs. Several new molecules have been identified as mu receptor G-protein-biased agonists, including oliceridine (TRV130), PZM21 and SR–17018. These compounds have provided preclinical data with apparent support for bias toward G proteins and the genetic premise of effective and safer analgesics. There are clinical data for oliceridine that have been very recently approved for short term intravenous use in hospitals and other controlled settings. While these data are compelling and provide a potential new pathway-based target for drug discovery, a simpler explanation for the behavior of these biased agonists revolves around differences in intrinsic activity. A highly detailed study comparing oliceridine, PZM21 and SR–17018 (among others) in a range of assays showed that these molecules behave as partial agonists. Moreover, there was a correlation between their therapeutic indices and their efficacies, but not their bias factors. If there is amplification of G-protein, but not arrestin pathways, then agonists with reduced efficacy would show high levels of activity at G-protein and low or absent activity at arrestin; offering analgesia with reduced side effects or ‘apparent bias’. Overall, the current data suggests—and we support—caution in ascribing biased agonism to reduced-side-effect profiles for mu-agonist analgesics.
Collapse
Affiliation(s)
- Joaquim Azevedo Neto
- Department of Biomedical and Specialty Surgical Sciences, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy; (J.A.N.); (G.C.)
| | - Anna Costanzini
- Department of Morphology, Surgery, Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (A.C.); (R.D.G.)
| | - Roberto De Giorgio
- Department of Morphology, Surgery, Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (A.C.); (R.D.G.)
| | - David G. Lambert
- Department of Cardiovascular Sciences, Anesthesia, Critical Care and Pain Management, University of Leicester, Leicester LE1 7RH, UK;
| | - Chiara Ruzza
- Department of Biomedical and Specialty Surgical Sciences, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy; (J.A.N.); (G.C.)
- Technopole of Ferrara, LTTA Laboratory for Advanced Therapies, 44122 Ferrara, Italy
- Correspondence:
| | - Girolamo Calò
- Department of Biomedical and Specialty Surgical Sciences, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy; (J.A.N.); (G.C.)
| |
Collapse
|
41
|
Krüll J, Fehler SK, Hofmann L, Nebel N, Maschauer S, Prante O, Gmeiner P, Lanig H, Hübner H, Heinrich MR. Synthesis, Radiosynthesis and Biological Evaluation of Buprenorphine-Derived Phenylazocarboxamides as Novel μ-Opioid Receptor Ligands. ChemMedChem 2020; 15:1175-1186. [PMID: 32378310 PMCID: PMC7383964 DOI: 10.1002/cmdc.202000180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Indexed: 12/12/2022]
Abstract
Targeted structural modifications have led to a novel type of buprenorphine-derived opioid receptor ligand displaying an improved selectivity profile for the μ-OR subtype. On this basis, it is shown that phenylazocarboxamides may serve as useful bioisosteric replacements for the widely occurring cinnamide units, without loss of OR binding affinity or subtype selectivity. This study further includes functional experiments pointing to weak partial agonist properties of the novel μ-OR ligands, as well as docking and metabolism experiments. Finally, the unique bifunctional character of phenylazocarboxylates, herein serving as precursors for the azocarboxamide subunit, was exploited to demonstrate the accessibility of an 18 F-fluorinated analogue.
Collapse
Affiliation(s)
- Jasmin Krüll
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Stefanie K. Fehler
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Laura Hofmann
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Natascha Nebel
- Department of Nuclear MedicineMolecular Imaging and RadiochemistryFriedrich-Alexander-Universität Erlangen-NürnbergSchwabachanlage 1291054ErlangenGermany
| | - Simone Maschauer
- Department of Nuclear MedicineMolecular Imaging and RadiochemistryFriedrich-Alexander-Universität Erlangen-NürnbergSchwabachanlage 1291054ErlangenGermany
| | - Olaf Prante
- Department of Nuclear MedicineMolecular Imaging and RadiochemistryFriedrich-Alexander-Universität Erlangen-NürnbergSchwabachanlage 1291054ErlangenGermany
| | - Peter Gmeiner
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Harald Lanig
- Central Institute for Scientific Computing (ZISC)Friedrich-Alexander-Universität Erlangen-NürnbergMartensstr. 5a91058ErlangenGermany
| | - Harald Hübner
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Markus R. Heinrich
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| |
Collapse
|
42
|
Abstract
Buprenorphine has not only had an interdisciplinary impact on our understanding of key neuroscience topics like opioid pharmacology, pain signaling, and reward processing but has also been a key influence in changing the way that substance use disorders are approached in modern medical systems. From its leading role in expanding outpatient treatment of opioid use disorders to its continued influence on research into next-generation analgesics, buprenorphine has been a continuous player in the ever-evolving societal perception of opioids and substance use disorder. To provide a multifaceted account on the enormous diversity of areas where this molecule has made an impact, this article discusses buprenorphine's chemical properties, synthesis and development, pharmacology, adverse effects, manufacturing information, and historical place in the field of chemical neuroscience.
Collapse
Affiliation(s)
- Jillian L. Kyzer
- University of Wisconsin-Madison, School of Pharmacy, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Cody J. Wenthur
- University of Wisconsin-Madison, School of Pharmacy, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| |
Collapse
|
43
|
Baptista-Hon DT, Smith M, Singleton S, Antonides LH, Nic Daeid N, McKenzie C, Hales TG. Activation of μ-opioid receptors by MT-45 (1-cyclohexyl-4-(1,2-diphenylethyl)piperazine) and its fluorinated derivatives. Br J Pharmacol 2020; 177:3436-3448. [PMID: 32246840 PMCID: PMC7348096 DOI: 10.1111/bph.15064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 03/03/2020] [Accepted: 03/21/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE A fluorinated derivative (2F-MT-45) of the synthetic μ-opioid receptor agonist MT-45 (1-cyclohexyl-4-(1,2-diphenylethyl)piperazine) was recently identified in a seized illicit tablet. While MT-45 is a Class A drug, banned in a number of countries, nothing is known about the pharmacology of 2F-MT-45. This study compares the pharmacology of MT-45, its fluorinated derivatives and two of its metabolites. EXPERIMENTAL APPROACH We used a β-arrestin2 recruitment assay in CHO cells stably expressing μ receptors to quantify the apparent potencies and efficacies of known (MT-45, morphine, fentanyl and DAMGO) and potential agonists. In addition, the GloSensor protein was transiently expressed to quantify changes in cAMP levels. We measured Ca2+ to investigate whether MT-45 and its metabolites have effects on GluN1/N2A NMDA receptors stably expressed in Ltk- cells. KEY RESULTS The fluorinated MT-45 derivatives have higher apparent potencies (2F-MT-45: 42 nM) than MT-45 (1.3 μM) for inhibition of cAMP accumulation and β-arrestin2 recruitment (2F-MT-45: 196 nM; MT-45: 23.1 μM). While MT-45 and 2F-MT-45 are poor recruiters of β-arrestin2, they have similar efficacies for reducing cAMP levels as DAMGO. Two MT-45 metabolites displayed negligible potencies as μ receptor agonists, but one, 1,2-diphenylethylpiperazine, inhibited the NMDA receptor with an IC50 of 29 μM. CONCLUSION AND IMPLICATIONS Fluorinated derivatives of MT-45 are potent μ receptor agonists and this may pose a danger to illicit opioid users. Inhibition of NMDA receptors by a metabolite of MT-45 may contribute to the reported dissociative effects.
Collapse
Affiliation(s)
- Daniel T Baptista-Hon
- Institute of Academic Anaesthesia, Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Mark Smith
- Institute of Academic Anaesthesia, Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Samuel Singleton
- Institute of Academic Anaesthesia, Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Lysbeth H Antonides
- Leverhulme Research Centre for Forensic Science, School of Science and Engineering, University of Dundee, Dundee, UK
| | - Niamh Nic Daeid
- Leverhulme Research Centre for Forensic Science, School of Science and Engineering, University of Dundee, Dundee, UK
| | - Craig McKenzie
- Leverhulme Research Centre for Forensic Science, School of Science and Engineering, University of Dundee, Dundee, UK
| | - Tim G Hales
- Institute of Academic Anaesthesia, Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
44
|
Piekielna-Ciesielska J, Wtorek K, Janecka A. Biased Agonism as an Emerging Strategy in the Search for Better Opioid Analgesics. Curr Med Chem 2020; 27:1562-1575. [PMID: 31057099 DOI: 10.2174/0929867326666190506103124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 12/12/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022]
Abstract
Morphine and related drugs that act through activating opioid receptors are the most effective analgesics for the relief of severe pain. They have been used for decades, despite the range of unwanted side effects that they produce, as no alternative has been found so far. The major goal of opioid research is to understand the mechanism of action of opioid receptor agonists and to improve the therapeutic utility of opioid drugs. In the search for safer and more potent analgesics, analogs with mixed opioid receptor profile gained a lot of interest. However, recently the concept of biased agonism, that highlights the fact that some ligands are able to differentially activate receptor downstream pathways, became a new approach in the design of novel drug candidates for clinical application. In this review, we summarize current knowledge on the development of opioid ligands of peptide and nonpeptide structure, showing how much opioid pharmacology evolved in recent years.
Collapse
Affiliation(s)
| | - Karol Wtorek
- Department of Biomolecular Chemistry, Medical University, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University, Mazowiecka 6/8, 92-215 Lodz, Poland
| |
Collapse
|
45
|
Duron DI, Lei W, Barker NK, Stine C, Mishra S, Blagg BSJ, Langlais PR, Streicher JM. Inhibition of Hsp90 in the spinal cord enhances the antinociceptive effects of morphine by activating an ERK-RSK pathway. Sci Signal 2020; 13:13/630/eaaz1854. [PMID: 32371496 DOI: 10.1126/scisignal.aaz1854] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Morphine and other opioids are commonly used to treat pain despite their numerous adverse side effects. Modulating μ-opioid receptor (MOR) signaling is one way to potentially improve opioid therapy. In mice, the chaperone protein Hsp90 mediates MOR signaling within the brain. Here, we found that inhibiting Hsp90 specifically in the spinal cord enhanced the antinociceptive effects of morphine in mice. Intrathecal, but not systemic, administration of the Hsp90 inhibitors 17-AAG or KU-32 amplified the effects of morphine in suppressing sensitivity to both thermal and mechanical stimuli in mice. Hsp90 inhibition enabled opioid-induced phosphorylation of the kinase ERK and increased abundance of the kinase RSK in the dorsal horns of the spinal cord, which are heavily populated with primary afferent sensory neurons. The additive effects of Hsp90 inhibition were abolished upon intrathecal inhibition of ERK, RSK, or protein synthesis. This mechanism downstream of MOR, localized to the spinal cord and repressed by Hsp90, may potentially be used to enhance the efficacy and presumably decrease the side effects of opioid therapy.
Collapse
Affiliation(s)
- David I Duron
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Wei Lei
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Natalie K Barker
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Carrie Stine
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Sanket Mishra
- Department of Chemistry and Biochemistry, College of Science, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, College of Science, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Paul R Langlais
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - John M Streicher
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
46
|
Hedegaard MA, Holst B. The Complex Signaling Pathways of the Ghrelin Receptor. Endocrinology 2020; 161:5734640. [PMID: 32049280 DOI: 10.1210/endocr/bqaa020] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/10/2020] [Indexed: 12/13/2022]
Abstract
The ghrelin receptor (GhrR) is known for its strong orexigenic effects in pharmacological doses and has long been considered as a promising target for the treatment of obesity. Several antagonists have been developed to decrease the orexigenic signaling, but none of these have been approved for the treatment of obesity because of adverse effects and lack of efficacy. Heterodimerization and biased signaling are important concepts for G-protein coupled receptor (GPCR) signaling, and the influence of these aspects on the GhrR may be important for feeding behavior and obesity. GhrR has been described to heterodimerize with other GPCRs, such as the dopamine receptors 1 and 2, leading to a modulation of the signaling properties of both dimerization partners. Another complicating factor of GhrR-mediated signaling is its ability to activate several different signaling pathways on ligand stimulation. Importantly, some ligands have shown to be "biased" or "functionally selective," implying that the ligand favors a particular signaling pathway. These unique signaling properties could have a sizeable impact on the physiological functions of the GhrR system. Importantly, heterodimerization may explain why the GhrR is expressed in areas of the brain that are difficult for peptide ligands to access. One possibility is that the purpose of GhrR expression is to modulate the function of other receptors in addition to merely being independently activated. We suggest that a deeper understanding of the signaling properties of the GhrR will facilitate future drug discovery in the areas of obesity and weight management.
Collapse
Affiliation(s)
- Morten Adler Hedegaard
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Gillis A, Gondin AB, Kliewer A, Sanchez J, Lim HD, Alamein C, Manandhar P, Santiago M, Fritzwanker S, Schmiedel F, Katte TA, Reekie T, Grimsey NL, Kassiou M, Kellam B, Krasel C, Halls ML, Connor M, Lane JR, Schulz S, Christie MJ, Canals M. Low intrinsic efficacy for G protein activation can explain the improved side effect profiles of new opioid agonists. Sci Signal 2020; 13:13/625/eaaz3140. [PMID: 32234959 DOI: 10.1126/scisignal.aaz3140] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Biased agonism at G protein-coupled receptors describes the phenomenon whereby some drugs can activate some downstream signaling activities to the relative exclusion of others. Descriptions of biased agonism focusing on the differential engagement of G proteins versus β-arrestins are commonly limited by the small response windows obtained in pathways that are not amplified or are less effectively coupled to receptor engagement, such as β-arrestin recruitment. At the μ-opioid receptor (MOR), G protein-biased ligands have been proposed to induce less constipation and respiratory depressant side effects than opioids commonly used to treat pain. However, it is unclear whether these improved safety profiles are due to a reduction in β-arrestin-mediated signaling or, alternatively, to their low intrinsic efficacy in all signaling pathways. Here, we systematically evaluated the most recent and promising MOR-biased ligands and assessed their pharmacological profile against existing opioid analgesics in assays not confounded by limited signal windows. We found that oliceridine, PZM21, and SR-17018 had low intrinsic efficacy. We also demonstrated a strong correlation between measures of efficacy for receptor activation, G protein coupling, and β-arrestin recruitment for all tested ligands. By measuring the antinociceptive and respiratory depressant effects of these ligands, we showed that the low intrinsic efficacy of opioid ligands can explain an improved side effect profile. Our results suggest a possible alternative mechanism underlying the improved therapeutic windows described for new opioid ligands, which should be taken into account for future descriptions of ligand action at this important therapeutic target.
Collapse
Affiliation(s)
- Alexander Gillis
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, NSW 2006, Australia
| | - Arisbel B Gondin
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, VIC 3052, Australia
| | - Andrea Kliewer
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
| | - Julie Sanchez
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, VIC 3052, Australia.,Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Midlands, UK
| | - Herman D Lim
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, VIC 3052, Australia
| | - Claudia Alamein
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, NSW 2006, Australia
| | - Preeti Manandhar
- Department of Biomedical Sciences, Macquarie University, NSW 2109, Australia
| | - Marina Santiago
- Department of Biomedical Sciences, Macquarie University, NSW 2109, Australia
| | - Sebastian Fritzwanker
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
| | - Frank Schmiedel
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
| | - Timothy A Katte
- School of Chemistry, University of Sydney, NSW 2006, Australia
| | - Tristan Reekie
- School of Chemistry, University of Sydney, NSW 2006, Australia
| | - Natasha L Grimsey
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Michael Kassiou
- School of Chemistry, University of Sydney, NSW 2006, Australia
| | - Barrie Kellam
- School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Cornelius Krasel
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, VIC 3052, Australia
| | - Mark Connor
- Department of Biomedical Sciences, Macquarie University, NSW 2109, Australia
| | - J Robert Lane
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Midlands, UK
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
| | - Macdonald J Christie
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, NSW 2006, Australia.
| | - Meritxell Canals
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, VIC 3052, Australia. .,Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Midlands, UK
| |
Collapse
|
48
|
Madariaga-Mazón A, Hernández-Alvarado RB, Noriega-Colima KO, Osnaya-Hernández A, Martinez-Mayorga K. Toxicity of secondary metabolites. PHYSICAL SCIENCES REVIEWS 2019. [DOI: 10.1515/psr-2018-0116] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Abstract
Secondary metabolites, commonly referred to as natural products, are produced by living organisms and usually have pharmacological or biological activities. Secondary metabolites are the primary source for the discovery of new drugs. Furthermore, secondary metabolites are also used as food preservatives, biopesticides or as research tools. Although secondary metabolites are mainly used by their beneficial biological activity, the toxicity of some of them may limit their use. The toxicity assessment of any compound that is prone to be used in direct contact with human beings is of vital importance. There is a vast spectrum of experimental methods for toxicity evaluation, including in vitro and in vivo methodologies. In this work, we present an overview of the different sources, bioactivities, toxicities and chemical classification of secondary metabolites, followed by a sketch of the role of toxicity assessment in drug discovery and agrochemistry.
Collapse
|
49
|
Kudla L, Bugno R, Skupio U, Wiktorowska L, Solecki W, Wojtas A, Golembiowska K, Zádor F, Benyhe S, Buda S, Makuch W, Przewlocka B, Bojarski AJ, Przewlocki R. Functional characterization of a novel opioid, PZM21, and its effects on the behavioural responses to morphine. Br J Pharmacol 2019; 176:4434-4445. [PMID: 31347704 DOI: 10.1111/bph.14805] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE The concept of opioid ligands biased towards the G protein pathway with minimal recruitment of β-arrestin-2 is a promising approach for the development of novel, efficient, and potentially nonaddictive opioid therapeutics. A recently discovered biased μ-opioid receptor agonist, PZM21, showed analgesic effects with reduced side effects. Here, we aimed to further investigate the behavioural and biochemical properties of PZM21. EXPERIMENT APPROACH We evaluated antinociceptive effects of systemic and intrathecal PZM21 administration. Its addiction-like properties were determined using several behavioural approaches: conditioned place preference, locomotor sensitization, precipitated withdrawal, and self-administration. Also, effects of PZM21 on morphine-induced antinociception, tolerance, and reward were assessed. Effects of PZM21 on striatal release of monoamines were evaluated using brain microdialysis. KEY RESULTS PZM21 caused long-lasting dose-dependent antinociception. It did not induce reward- and reinforcement-related behaviour; however, its repeated administration led to antinociceptive tolerance and naloxone-precipitated withdrawal symptoms. Pretreatment with PZM21 enhanced morphine-induced antinociception and attenuated the expression of morphine reward. In comparison to morphine, PZM21 administration induced a moderate release of dopamine and a robust release of 5-HT in the striatum. CONCLUSIONS AND IMPLICATIONS PZM21 exhibited antinociceptive efficacy, without rewarding or reinforcing properties. However, its clinical application may be restricted, as it induces tolerance and withdrawal symptoms. Notably, its ability to diminish morphine reward implies that PZM21 may be useful in treatment of opioid use disorders.
Collapse
Affiliation(s)
- Lucja Kudla
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ryszard Bugno
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Urszula Skupio
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Lucja Wiktorowska
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wojciech Solecki
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Adam Wojtas
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Krystyna Golembiowska
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ferenc Zádor
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Sándor Benyhe
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Szymon Buda
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Wioletta Makuch
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Barbara Przewlocka
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Andrzej J Bojarski
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
50
|
Ma M, Li X, Tong K, Cheng J, Yu Z, Ren F, Zhong B, Shi W. Discovery of Biased Mu‐Opioid Receptor Agonists for the Treatment of Pain. ChemMedChem 2019; 15:155-161. [DOI: 10.1002/cmdc.201900575] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/06/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Mengjun Ma
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology & Toxicology 27 Tai-Ping Road Beijing 100850 China
| | - Xiang Li
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology & Toxicology 27 Tai-Ping Road Beijing 100850 China
| | - Kun Tong
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology & Toxicology 27 Tai-Ping Road Beijing 100850 China
| | - Jingchao Cheng
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology & Toxicology 27 Tai-Ping Road Beijing 100850 China
| | - Zixing Yu
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology & Toxicology 27 Tai-Ping Road Beijing 100850 China
| | - Fengxia Ren
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology & Toxicology 27 Tai-Ping Road Beijing 100850 China
| | - Bohua Zhong
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology & Toxicology 27 Tai-Ping Road Beijing 100850 China
| | - Weiguo Shi
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology & Toxicology 27 Tai-Ping Road Beijing 100850 China
| |
Collapse
|