1
|
Chen X, Ge Y, Shi W, Yang M, Zhou Q, Pan Y. Estro-Androgenic Disrupting Effects of Halogenated Disinfection Byproducts: A Comprehensive Evaluation and Comparison. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:20190-20200. [PMID: 39475525 DOI: 10.1021/acs.est.4c07223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2024]
Abstract
Drinking water halogenated disinfection byproducts (DBPs) have become an increasing health concern. However, the endocrine-disrupting effects of DBPs have not been well evaluated, and the limited available data have inhibited a comprehensive understanding of their health risks. In this study, a total of 43 DBPs were evaluated for their estro-androgenic effects using two types of human breast cancer cells. Among the tested DBPs, 16 exhibited estrogenic/antiestrogenic/androgenic/antiandrogenic effects, and the effects could be observed even at concentrations typically detected in drinking water. Iodinated and polyhalogenated DBPs generally showed higher effects than other species. For a broader comparison, DBP endocrine-disrupting effect data from this study and previous studies were summarized. It was found that the endocrine disruption efficacy of DBPs followed the rank order of iodinated > brominated > chlorinated species, and halophenolic DBPs were potential endocrine-disrupting compounds. Moreover, molecular docking results demonstrated that the binding of DBPs to estro-androgenic receptors was dominated by hydrophobic bonding, hydrogen bonding, halogen bonding, and van der Waals forces. The force strength and molecular volume were related to the magnitude of the estro-androgenic effects. Iodinated DBPs and polyhalogenated DBPs tended to have larger binding forces than other analogues and thus exhibited stronger effects.
Collapse
Affiliation(s)
- Xueyao Chen
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, Jiangsu, China
| | - Yaoming Ge
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, Jiangsu, China
| | - Wei Shi
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, Jiangsu, China
| | - Mengting Yang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Qing Zhou
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, Jiangsu, China
| | - Yang Pan
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, Jiangsu, China
| |
Collapse
|
2
|
Liu H, Zhang H, IJzerman AP, Guo D. The translational value of ligand-receptor binding kinetics in drug discovery. Br J Pharmacol 2024; 181:4117-4129. [PMID: 37705429 DOI: 10.1111/bph.16241] [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: 05/19/2023] [Revised: 07/27/2023] [Accepted: 09/01/2023] [Indexed: 09/15/2023] Open
Abstract
The translation of in vitro potency of a candidate drug, as determined by traditional pharmacology metrics (such as EC50/IC50 and KD/Ki values), to in vivo efficacy and safety is challenging. Residence time, which represents the duration of drug-target interaction, can be part of a more comprehensive understanding of the dynamic nature of drug-target interactions in vivo, thereby enabling better prediction of drug efficacy and safety. As a consequence, a prolonged residence time may help in achieving sustained pharmacological activity, while transient interactions with shorter residence times may be favourable for targets associated with side effects. Therefore, integration of residence time into the early stages of drug discovery and development has yielded a number of clinical candidates with promising in vivo efficacy and safety profiles. Insights from residence time research thus contribute to the translation of in vitro potency to in vivo efficacy and safety. Further research and advances in measuring and optimizing residence time will bring a much-needed addition to the drug discovery process and the development of safer and more effective drugs. In this review, we summarize recent research progress on residence time, highlighting its importance from a translational perspective.
Collapse
Affiliation(s)
- Hongli Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Haoran Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Adriaan P IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
3
|
Kosar M, Sarott RC, Sykes DA, Viray AEG, Vitale RM, Tomašević N, Li X, Ganzoni RLZ, Kicin B, Reichert L, Patej KJ, Gómez-Bouzó U, Guba W, McCormick PJ, Hua T, Gruber CW, Veprintsev DB, Frank JA, Grether U, Carreira EM. Flipping the GPCR Switch: Structure-Based Development of Selective Cannabinoid Receptor 2 Inverse Agonists. ACS CENTRAL SCIENCE 2024; 10:956-968. [PMID: 38799662 PMCID: PMC11117691 DOI: 10.1021/acscentsci.3c01461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 05/29/2024]
Abstract
We report a blueprint for the rational design of G protein coupled receptor (GPCR) ligands with a tailored functional response. The present study discloses the structure-based design of cannabinoid receptor type 2 (CB2R) selective inverse agonists (S)-1 and (R)-1, which were derived from privileged agonist HU-308 by introduction of a phenyl group at the gem-dimethylheptyl side chain. Epimer (R)-1 exhibits high affinity for CB2R with Kd = 39.1 nM and serves as a platform for the synthesis of a wide variety of probes. Notably, for the first time these fluorescent probes retain their inverse agonist functionality, high affinity, and selectivity for CB2R independent of linker and fluorophore substitution. Ligands (S)-1, (R)-1, and their derivatives act as inverse agonists in CB2R-mediated cAMP as well as G protein recruitment assays and do not trigger β-arrestin-receptor association. Furthermore, no receptor activation was detected in live cell ERK1/2 phosphorylation and Ca2+-release assays. Confocal fluorescence imaging experiments with (R)-7 (Alexa488) and (R)-9 (Alexa647) probes employing BV-2 microglial cells visualized CB2R expressed at endogenous levels. Finally, molecular dynamics simulations corroborate the initial docking data in which inverse agonists restrict movement of toggle switch Trp2586.48 and thereby stabilize CB2R in its inactive state.
Collapse
Affiliation(s)
- Miroslav Kosar
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Roman C. Sarott
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - David A. Sykes
- Faculty
of Medicine & Health Sciences, University
of Nottingham, Nottingham NG7 2UH, U.K.
- Centre
of Membrane Proteins and Receptors (COMPARE), University of Birmingham
and University of Nottingham, https://www.birmingham-nottingham.ac.uk/compare
| | - Alexander E. G. Viray
- Department
of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon 97239-3098, United States
| | - Rosa Maria Vitale
- Institute
of Biomolecular Chemistry, National Research
Council, Via Campi Flegrei
34, 80078 Pozzuoli, Italy
| | - Nataša Tomašević
- Center for
Physiology and Pharmacology, Medical University
of Vienna, Schwarzspanierstrasse
17, 1090 Vienna, Austria
| | - Xiaoting Li
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Rudolf L. Z. Ganzoni
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Bilal Kicin
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Lisa Reichert
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Kacper J. Patej
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Uxía Gómez-Bouzó
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Wolfgang Guba
- Roche
Pharma Research & Early Development, Roche Innovation Center Basel,
F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Peter J. McCormick
- Department
of Pharmacology and Therapeutics, University
of Liverpool, Ashton
Street, Liverpool L69 3GE, U.K.
| | - Tian Hua
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Christian W. Gruber
- Center for
Physiology and Pharmacology, Medical University
of Vienna, Schwarzspanierstrasse
17, 1090 Vienna, Austria
| | - Dmitry B. Veprintsev
- Faculty
of Medicine & Health Sciences, University
of Nottingham, Nottingham NG7 2UH, U.K.
- Centre
of Membrane Proteins and Receptors (COMPARE), University of Birmingham
and University of Nottingham, https://www.birmingham-nottingham.ac.uk/compare
| | - James A. Frank
- Department
of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon 97239-3098, United States
- Vollum
Institute, Oregon Health & Science University, Portland, Oregon 97239-3098, United States
| | - Uwe Grether
- Roche
Pharma Research & Early Development, Roche Innovation Center Basel,
F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Erick M. Carreira
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| |
Collapse
|
4
|
Vauquelin G, Maes D. Competition in drug binding and … the race to equilibrium. Fundam Clin Pharmacol 2023; 37:147-157. [PMID: 35981720 DOI: 10.1111/fcp.12824] [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: 03/31/2022] [Revised: 06/30/2022] [Accepted: 08/17/2022] [Indexed: 01/25/2023]
Abstract
Binding kinetics has become a popular topic in pharmacology due to its potential contribution to the selectivity and duration of drug action. Yet, the overall kinetic aspects of complex binding mechanisms are still merely described in terms of elaborate algebraic equations. Interestingly, it has been recommended some 10 years ago to examine such mechanisms in terms of binding fluxes instead of the conventional rate constants. Alike the velocity of product formation in enzymology, those fluxes refer to the velocity by which one target species converts into another one. Novel binding flux-based approaches are utilized to get a better visual insight into the "competition" between two drugs/ligands for a single target as well as between induced fit- and conformational selection pathways for a single ligand within a thermodynamic cycle. The present data were obtained by differential equation-based simulations. Early on, the ligand-binding steps "race" to equilibrium (i.e., when their forward and reverse fluxes are equal) at their individual pace. The overall/global equilibrium is only reached later on. For the competition association assays, this parting might produce a transient "overshoot" of one of the bound target species. A similar overshoot may also show up within a thermodynamic cycle and, at first glance, suggest that the induced fit pathway dominates. Yet, present findings show that under certain circumstances, it could rather be the other way round. Novel binding flux-based approaches offer visually attractive insights into crucial aspects of "complex" binding mechanisms under non-equilibrium conditions.
Collapse
Affiliation(s)
- Georges Vauquelin
- Department of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Dominique Maes
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
5
|
Grätz L, Müller C, Pegoli A, Schindler L, Bernhardt G, Littmann T. Insertion of Nanoluc into the Extracellular Loops as a Complementary Method To Establish BRET-Based Binding Assays for GPCRs. ACS Pharmacol Transl Sci 2022; 5:1142-1155. [PMID: 36407949 PMCID: PMC9667534 DOI: 10.1021/acsptsci.2c00162] [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: 08/15/2022] [Indexed: 11/13/2022]
Abstract
Luminescence-based techniques play an increasingly important role in all areas of biochemical research, including investigations on G protein-coupled receptors (GPCRs). One quite recent and popular addition has been made by introducing bioluminescence resonance energy transfer (BRET)-based binding assays for GPCRs, which are based on the fusion of nanoluciferase (Nluc) to the N-terminus of the receptor and the occurring energy transfer via BRET to a bound fluorescent ligand. However, being based on BRET, the technique is strongly dependent on the distance/orientation between the luciferase and the fluorescent ligand. Here we describe an alternative strategy to establish BRET-based binding assays for GPCRs, where the N-terminal fusion of Nluc did not result in functioning test systems with our fluorescent ligands (e.g., for the neuropeptide Y Y1 receptor (Y1R) and the neurotensin receptor type 1 (NTS1R)). Instead, we introduced Nluc into their second extracellular loop and we obtained binding data for the fluorescent ligands and reported standard ligands (in saturation and competition binding experiments, respectively) comparable to data from the literature. The strategy was transferred to the angiotensin II receptor type 1 (AT1R) and the M1 muscarinic acetylcholine receptor (M1R), which led to affinity estimates comparable to data from radioligand binding experiments. Additionally, an analysis of the binding kinetics of all fluorescent ligands at their respective target was performed using the newly described receptor/Nluc-constructs.
Collapse
Affiliation(s)
| | - Christoph Müller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | | | - Lisa Schindler
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Günther Bernhardt
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | | |
Collapse
|
6
|
Tahk MJ, Torp J, Ali MAS, Fishman D, Parts L, Grätz L, Müller C, Keller M, Veiksina S, Laasfeld T, Rinken A. Live-cell microscopy or fluorescence anisotropy with budded baculoviruses-which way to go with measuring ligand binding to M 4 muscarinic receptors? Open Biol 2022; 12:220019. [PMID: 35674179 PMCID: PMC9175271 DOI: 10.1098/rsob.220019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/27/2022] [Indexed: 01/04/2023] Open
Abstract
M4 muscarinic acetylcholine receptor is a G protein-coupled receptor (GPCR) that has been associated with alcohol and cocaine abuse, Alzheimer's disease, and schizophrenia which makes it an interesting drug target. For many GPCRs, the high-affinity fluorescence ligands have expanded the options for high-throughput screening of drug candidates and serve as useful tools in fundamental receptor research. Here, we explored two TAMRA-labelled fluorescence ligands, UR-MK342 and UR-CG072, for development of assays for studying ligand-binding properties to M4 receptor. Using budded baculovirus particles as M4 receptor preparation and fluorescence anisotropy method, we measured the affinities and binding kinetics of both fluorescence ligands. Using the fluorescence ligands as reporter probes, the binding affinities of unlabelled ligands could be determined. Based on these results, we took a step towards a more natural system and developed a method using live CHO-K1-hM4R cells and automated fluorescence microscopy suitable for the routine determination of unlabelled ligand affinities. For quantitative image analysis, we developed random forest and deep learning-based pipelines for cell segmentation. The pipelines were integrated into the user-friendly open-source Aparecium software. Both image analysis methods were suitable for measuring fluorescence ligand saturation binding and kinetics as well as for screening binding affinities of unlabelled ligands.
Collapse
Affiliation(s)
- Maris-Johanna Tahk
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Jane Torp
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Mohammed A. S. Ali
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
| | - Dmytro Fishman
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
| | - Leopold Parts
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
| | - Lukas Grätz
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Christoph Müller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Santa Veiksina
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Tõnis Laasfeld
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
| | - Ago Rinken
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| |
Collapse
|
7
|
Penna E, Niso M, Podlewska S, Volpicelli F, Crispino M, Perrone-Capano C, Bojarski AJ, Lacivita E, Leopoldo M. In Vitro and In Silico Analysis of the Residence Time of Serotonin 5-HT 7 Receptor Ligands with Arylpiperazine Structure: A Structure-Kinetics Relationship Study. ACS Chem Neurosci 2022; 13:497-509. [PMID: 35099177 DOI: 10.1021/acschemneuro.1c00710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
During the last decade, the kinetics of drug-target interaction has received increasing attention as an important pharmacological parameter in the drug development process. Several studies have suggested that the lipophilicity of a molecule can play an important role. To date, this aspect has been studied for several G protein-coupled receptors (GPCRs) ligands but not for the 5-HT7 receptor (5-HT7R), a GPCR proposed as a valid therapeutic target in neurodevelopmental and neuropsychiatric disorders associated with abnormal neuronal connectivity. In this study, we report on structure-kinetics relationships of a set of arylpiperazine-based 5-HT7R ligands. We found that it is not the overall lipophilicity of the molecule that influences drug-target interaction kinetics but rather the position of polar groups within the molecule. Next, we performed a combination of molecular docking studies and molecular dynamics simulations to gain insights into structure-kinetics relationships. These studies did not suggest specific contact patterns between the ligands and the receptor-binding site as determinants for compounds kinetics. Finally, we compared the abilities of two 5-HT7R agonists with similar receptor-binding affinities and different residence times to stimulate the 5-HT7R-mediated neurite outgrowth in mouse neuronal primary cultures and found that the compounds induced the effect with different timing. This study provides the first insights into the binding kinetics of arylpiperazine-based 5-HT7R ligands that can be helpful to design new 5-HT7R ligands with fine-tuning of the kinetic profile.
Collapse
Affiliation(s)
- Eduardo Penna
- Department of Biology, University of Naples Federico II, via Cintia 26, 80126 Naples, Italy
- Biofordrug srl, via Dante 99, 70019 Triggiano (Bari), Italy
| | - Mauro Niso
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona 4, 70125 Bari, Italy
| | - Sabina Podlewska
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland
| | - Floriana Volpicelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, via Domenico Montesano 49, 80131 Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, via Cintia 26, 80126 Naples, Italy
| | - Carla Perrone-Capano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, via Domenico Montesano 49, 80131 Naples, Italy
- Institute of Genetics and Biophysics “Adriano Buzzati Traverso”, National Research Council (CNR), via Pietro Castellino 111, 80131 Naples, Italy
| | - Andrzej J. Bojarski
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland
| | - Enza Lacivita
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona 4, 70125 Bari, Italy
| | - Marcello Leopoldo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona 4, 70125 Bari, Italy
| |
Collapse
|
8
|
Long-Timescale Simulations Revealed Critical Non-Conserved Residues of Phosphodiesterases Affecting Selectivity of BAY60-7550. Comput Struct Biotechnol J 2022; 20:5136-5149. [PMID: 36187927 PMCID: PMC9508422 DOI: 10.1016/j.csbj.2022.09.013] [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] [Received: 06/11/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
A major obstacle of the selective inhibitor design for specific human phosphodiesterase (PDE) is that highly conserved catalytic pockets are difficult to be distinguished by inhibitor molecules. To overcome this, a feasible path is to understand the molecular determinants underlying the selectivity of current inhibitors. BAY60-7550 (BAY for short; IC50 = 4.7 nM) is a highly selective inhibitor targeting PDE2A which is a dual-specificity PDE and an attractive target for therapeutic intervention of the central nervous system (CNS) disorders. Recent studies suggest that molecular determinants may be in binding processes of BAY. However, a detailed understanding of these processes are still lacking. To explore these processes, High-Throughput Molecular Dynamics (HTMD) simulations were performed to reproduce the spontaneous association of BAY with catalytic pockets of 4 PDE isoforms; Ligand Gaussian Accelerated Molecular Dynamics (LiGaMD) simulations were performed to reproduce the unbinding-rebinding processes of FKG and 10.13039/100016266MC2, two pyrazolopyrimidinone PDE2A selective inhibitors, in the PDE2A system. The produced molecular trajectories were analyzed by the Markov state model (MSM) and the molecular mechanics/generalized Born surface area (MM/GBSA). The results showed that the non-covalent interactions between the non-conserved residues and BAY, especially the hydrogen bonds, determined the unique binding pathways of BAY on the surface of PDE2A. These pathways were different from those of BAY on the surface of the other three PDE isoforms and the binding pathways of the other two PDE2A inhibitors in PDE2A systems. These differences were ultimately reflected in the high selectivity of this inhibitor for PDE2A. As a result, this study demonstrates the critical role of the binding processes in the selectivity of BAY, and also identifies the key non-conserved residues affecting the binding processes of BAY. Thus, this study provides a new perspective and data support for the further development of BAY-derived inhibitors targeting PDE2A.
Collapse
|
9
|
Duarte DA, Parreiras-E-Silva LT, Oliveira EB, Bouvier M, Costa-Neto CM. Angiotensin II Type 1 Receptor Tachyphylaxis Is Defined by Agonist Residence Time. Hypertension 2021; 79:115-125. [PMID: 34739768 DOI: 10.1161/hypertensionaha.121.17977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Several GPCRs (G-protein-coupled receptors) have been reported to exhibit tachyphylaxis, which is an acute loss of functional receptor response after repeated stimuli with an agonist. GPCRs are important clinical targets for a wide range of disorders. Therefore, elucidation of the ligand features that contribute to receptor tachyphylaxis and signaling events underlying this phenomenon is important for drug discovery and development. In this study, we examined the role of ligand-binding kinetics in the tachyphylaxis of AT1R (angiotensin II type 1 receptor) using bioluminescence resonance energy transfer assays to monitor signaling events under both kinetic and equilibrium conditions. We investigated AT1R signal transduction and translocation promoted by the endogenous tachyphylactic agonist Ang II (angiotensin II) and its analogs, described previously for inducing reduced receptor tachyphylaxis. Estimation of binding kinetic parameters of the ligands revealed that the residence time of Ang II was higher than that of the analogs, resulting in more sustained Gq protein activation and recruitment of β-arrestin than that promoted by the analogs. Furthermore, we observed that Ang II led to more sustained internalization of the receptor, thereby retarding its recycling to the plasma membrane and preventing further receptor responses. These results show that the apparent lack of tachyphylaxis in the studied analogs resulted from their short residence time at the AT1R. In addition, our data highlight the relevance of complete characterization of novel GPCR drug candidates, taking into account their receptor binding kinetics as well.
Collapse
Affiliation(s)
- Diego A Duarte
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil (D.A.D., L.T.P.-e.-S., E.B.O., C.M.C.-N.)
| | - Lucas T Parreiras-E-Silva
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil (D.A.D., L.T.P.-e.-S., E.B.O., C.M.C.-N.)
| | - Eduardo B Oliveira
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil (D.A.D., L.T.P.-e.-S., E.B.O., C.M.C.-N.)
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, QC, Canada (M.B.)
| | - Claudio M Costa-Neto
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil (D.A.D., L.T.P.-e.-S., E.B.O., C.M.C.-N.)
| |
Collapse
|
10
|
Vauquelin G, Maes D. Induced fit versus conformational selection: From rate constants to fluxes… and back to rate constants. Pharmacol Res Perspect 2021; 9:e00847. [PMID: 34459109 PMCID: PMC8404059 DOI: 10.1002/prp2.847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/07/2021] [Indexed: 12/30/2022] Open
Abstract
Induced fit- (IF) and conformational selection (CS) binding mechanisms have long been regarded to be mutually exclusive. Yet, they are now increasingly considered to produce the final ligand-target complex alongside within a thermodynamic cycle. This viewpoint benefited from the introduction of binding fluxes as a tool for analyzing the overall behavior of such cycle. This study aims to provide more vivid and applicable insights into this emerging field. In this respect, combining differential equation- based simulations and hitherto little explored alternative modes of calculation provide concordant information about the intricate workings of such cycle. In line with previous reports, we observe that the relative contribution of IF increases with the ligand concentration at equilibrium. Yet the baseline contribution may vary from one case to another and simulations as well as calculations show that this parameter is essentially regulated by the dissociation rate of both pathways. Closer attention should be paid to how the contributions of IF and CS compare at physiologically relevant drug/ligand concentrations. To this end, a simple equation discloses how changing a limited set of "microscopic" rate constants can extend the concentration range at which CS contributes most effectively. Finally, it could also be beneficial to extend the utilization of flux- based approaches to more physiologically relevant time scales and alternative binding models.
Collapse
Affiliation(s)
- Georges Vauquelin
- Department Molecular and Biochemical PharmacologyVrije Universiteit BrusselBrusselsBelgium
| | - Dominique Maes
- Structural Biology BrusselsVrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
11
|
Wang Z, Bosma R, Kuhne S, van den Bor J, Garabitian W, Vischer HF, Wijtmans M, Leurs R, de Esch IJ. Exploring the Effect of Cyclization of Histamine H 1 Receptor Antagonists on Ligand Binding Kinetics. ACS OMEGA 2021; 6:12755-12768. [PMID: 34056427 PMCID: PMC8154229 DOI: 10.1021/acsomega.0c06358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/16/2021] [Indexed: 06/12/2023]
Abstract
There is an increasing interest in guiding hit optimization by considering the target binding kinetics of ligands. However, compared to conventional structure-activity relationships, structure-kinetics relationships have not been as thoroughly explored, even for well-studied archetypical drug targets such as the histamine H1 receptor (H1R), a member of the family A G-protein coupled receptor. In this study, we show that the binding kinetics of H1R antagonists at the H1R is dependent on the cyclicity of both the aromatic head group and the amine moiety of H1R ligands, the chemotypes that are characteristic for the first-generation H1R antagonists. Fusing the two aromatic rings of H1R ligands into one tricyclic aromatic head group prolongs the H1R residence time for benchmark H1R ligands as well as for tailored synthetic analogues. The effect of constraining the aromatic rings and the basic amines is systematically explored, leading to a coherent series and detailed discussions of structure-kinetics relationships. This study shows that cyclicity has a pronounced effect on the binding kinetics.
Collapse
Affiliation(s)
| | | | | | - Jelle van den Bor
- Amsterdam Institute of Molecular
and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty
of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Wrej Garabitian
- Amsterdam Institute of Molecular
and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty
of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Henry F. Vischer
- Amsterdam Institute of Molecular
and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty
of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Maikel Wijtmans
- Amsterdam Institute of Molecular
and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty
of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rob Leurs
- Amsterdam Institute of Molecular
and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty
of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J.P. de Esch
- Amsterdam Institute of Molecular
and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty
of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
12
|
Comeo E, Trinh P, Nguyen AT, Nowell CJ, Kindon ND, Soave M, Stoddart LA, White JM, Hill SJ, Kellam B, Halls ML, May LT, Scammells PJ. Development and Application of Subtype-Selective Fluorescent Antagonists for the Study of the Human Adenosine A 1 Receptor in Living Cells. J Med Chem 2021; 64:6670-6695. [PMID: 33724031 DOI: 10.1021/acs.jmedchem.0c02067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The adenosine A1 receptor (A1AR) is a G-protein-coupled receptor (GPCR) that provides important therapeutic opportunities for a number of conditions including congestive heart failure, tachycardia, and neuropathic pain. The development of A1AR-selective fluorescent ligands will enhance our understanding of the subcellular mechanisms underlying A1AR pharmacology facilitating the development of more efficacious and selective therapies. Herein, we report the design, synthesis, and application of a novel series of A1AR-selective fluorescent probes based on 8-functionalized bicyclo[2.2.2]octylxanthine and 3-functionalized 8-(adamant-1-yl) xanthine scaffolds. These fluorescent conjugates allowed quantification of kinetic and equilibrium ligand binding parameters using NanoBRET and visualization of specific receptor distribution patterns in living cells by confocal imaging and total internal reflection fluorescence (TIRF) microscopy. As such, the novel A1AR-selective fluorescent antagonists described herein can be applied in conjunction with a series of fluorescence-based techniques to foster understanding of A1AR molecular pharmacology and signaling in living cells.
Collapse
Affiliation(s)
- Eleonora Comeo
- Medicinal Chemistry, Monash University, Parkville, Victoria 3052, Australia.,Division of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, B15 2TT and University of Nottingham, Birmingham NG7 2UH, United Kingdom
| | - Phuc Trinh
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Anh T Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Nicholas D Kindon
- Division of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, B15 2TT and University of Nottingham, Birmingham NG7 2UH, United Kingdom
| | - Mark Soave
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, B15 2TT and University of Nottingham, Birmingham NG7 2UH, United Kingdom
| | - Leigh A Stoddart
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, B15 2TT and University of Nottingham, Birmingham NG7 2UH, United Kingdom
| | - Jonathan M White
- School of Chemistry and the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, B15 2TT and University of Nottingham, Birmingham NG7 2UH, United Kingdom
| | - Barrie Kellam
- Division of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, B15 2TT and University of Nottingham, Birmingham NG7 2UH, United Kingdom
| | - Michelle L Halls
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Peter J Scammells
- Medicinal Chemistry, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
13
|
Abid MSR, Mousavi S, Checco JW. Identifying Receptors for Neuropeptides and Peptide Hormones: Challenges and Recent Progress. ACS Chem Biol 2021; 16:251-263. [PMID: 33539706 DOI: 10.1021/acschembio.0c00950] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intercellular signaling events mediated by neuropeptides and peptide hormones represent important targets for both basic science and drug discovery. For many bioactive peptides, the protein receptors that transmit information across the receiving cell membrane are not known, severely limiting these signaling pathways as potential therapeutic targets. Identifying the receptor(s) for a given peptide of interest is complicated by several factors. Most notably, cell-cell signaling peptides are generated through dynamic biosynthetic pathways, can act on many different families of receptor proteins, and can participate in complex ligand-receptor interactions that extend beyond a simple one-to-one archetype. Here, we discuss recent methodological advances to identify signaling partners for bioactive peptides. Recent efforts have centered on methods to identify candidate receptors via transcript expression, methods to match peptide-receptor pairs through high throughput screening, and methods to capture direct ligand-receptor interactions using chemical probes. Future applications of the receptor identification approaches discussed here, as well as technical advancements to address their limitations, promise to lead to a greater understanding of how cells communicate to deliver complex physiologies. Importantly, such advancements will likely provide novel targets for the treatment of human diseases within the central nervous and endocrine systems.
Collapse
Affiliation(s)
- Md Shadman Ridwan Abid
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Somayeh Mousavi
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - James W. Checco
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| |
Collapse
|
14
|
Veiksina S, Tahk MJ, Laasfeld T, Link R, Kopanchuk S, Rinken A. Fluorescence Anisotropy-Based Assay for Characterization of Ligand Binding Dynamics to GPCRs: The Case of Cy3B-Labeled Ligands Binding to MC 4 Receptors in Budded Baculoviruses. Methods Mol Biol 2021; 2268:119-136. [PMID: 34085265 DOI: 10.1007/978-1-0716-1221-7_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
During the past decade, fluorescence methods have become valuable tools for characterizing ligand binding to G protein-coupled receptors (GPCRs). However, only a few of the assays enable studying wild-type receptors and monitor the ligand binding in real time. One of the approaches that is inherently suitable for this purpose is the fluorescence anisotropy (FA) assay. In the FA assay, the change of ligand's rotational freedom connected with its binding to the receptor can be monitored with a conventional fluorescence plate reader equipped with suitable optical filters. To achieve the high receptor concentration required for the assay and the low autofluorescence levels essential for reliable results, budded baculoviruses that display GPCRs on their surfaces can be used. The monitoring process generates a substantial amount of kinetic data, which is usually stored as a proprietary file format limiting the flexibility of data analysis. To solve this problem, we propose the use of the data curation software Aparecium ( http://gpcr.ut.ee/aparecium.html ), which integrates experimental data with metadata in a Minimum Information for Data Analysis in Systems Biology (MIDAS) format. Aparecium enables data export to different software packages for fitting to suitable kinetic or equilibrium models. A combination of the FA assay with the novel data analysis strategy is suitable for screening new active compounds, but also for modeling complex systems of ligand binding to GPCRs. We present the proposed approach using different fluorescent probes and assay types to characterize ligand binding to melanocortin 4 (MC4) receptor.
Collapse
Affiliation(s)
- Santa Veiksina
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | | | - Tõnis Laasfeld
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Reet Link
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | | | - Ago Rinken
- Institute of Chemistry, University of Tartu, Tartu, Estonia.
| |
Collapse
|
15
|
BRET- and fluorescence anisotropy-based assays for real-time monitoring of ligand binding to M 2 muscarinic acetylcholine receptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118930. [PMID: 33347921 DOI: 10.1016/j.bbamcr.2020.118930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/26/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022]
Abstract
BRET and fluorescence anisotropy (FA) are two fluorescence-based techniques used for the characterization of ligand binding to G protein-coupled receptors (GPCRs) and both allow monitoring of ligand binding in real time. In this study, we present the first direct comparison of BRET-based and FA-based binding assays using the human M2 muscarinic acetylcholine receptor (M2R) and two TAMRA (5-carboxytetramethylrhodamine)-labeled fluorescent ligands as a model system. The determined fluorescent ligand affinities from both assays were in good agreement with results obtained from radioligand competition binding experiments. The assays yielded real-time kinetic binding data revealing differences in the mechanism of binding for the investigated fluorescent probes. Furthermore, the investigation of various unlabeled M2R ligands yielded pharmacological profiles in accordance with earlier reported data. Taken together, this study showed that BRET- and FA-based binding assays represent valuable alternatives to radioactivity-based methods for screening purposes and for a precise characterization of binding kinetics supporting the exploration of binding mechanisms.
Collapse
|
16
|
New small molecule fluorescent probes for G protein-coupled receptors: valuable tools for drug discovery. Future Med Chem 2020; 13:63-90. [PMID: 33319586 DOI: 10.4155/fmc-2019-0327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are essential signaling proteins and tractable therapeutic targets. To develop new drug candidates, GPCR drug discovery programs require versatile, sensitive pharmacological tools for ligand binding and compound screening. With the availability of new imaging modalities and proximity-based ligand binding technologies, fluorescent ligands offer many advantages and are increasingly being used, yet labeling small molecules remains considerably more challenging relative to peptides. Focusing on recent fluorescent small molecule studies for family A GPCRs, this review addresses some of the key challenges, synthesis approaches and structure-activity relationship considerations, and discusses advantages of using high-resolution GPCR structures to inform conjugation strategies. While no single approach guarantees successful labeling without loss of affinity or selectivity, the choice of fluorophore, linker type and site of attachment have proved to be critical factors that can significantly affect their utility in drug discovery programs, and as discussed, can sometimes lead to very unexpected results.
Collapse
|
17
|
Höring C, Seibel U, Tropmann K, Grätz L, Mönnich D, Pitzl S, Bernhardt G, Pockes S, Strasser A. A Dynamic, Split-Luciferase-Based Mini-G Protein Sensor to Functionally Characterize Ligands at All Four Histamine Receptor Subtypes. Int J Mol Sci 2020; 21:ijms21228440. [PMID: 33182741 PMCID: PMC7698210 DOI: 10.3390/ijms21228440] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 02/06/2023] Open
Abstract
In drug discovery, assays with proximal readout are of great importance to study target-specific effects of potential drug candidates. In the field of G protein-coupled receptors (GPCRs), the determination of GPCR-G protein interactions and G protein activation by means of radiolabeled GTP analogs ([35S]GTPγS, [γ-32P]GTP) has widely been used for this purpose. Since we were repeatedly faced with insufficient quality of radiolabeled nucleotides, there was a requirement to implement a novel proximal functional assay for the routine characterization of putative histamine receptor ligands. We applied the split-NanoLuc to the four histamine receptor subtypes (H1R, H2R, H3R, H4R) and recently engineered minimal G (mini-G) proteins. Using this method, the functional response upon receptor activation was monitored in real-time and the four mini-G sensors were evaluated by investigating selected standard (inverse) agonists and antagonists. All potencies and efficacies of the studied ligands were in concordance with literature data. Further, we demonstrated a significant positive correlation of the signal amplitude and the mini-G protein expression level in the case of the H2R, but not for the H1R or the H3R. The pEC50 values of histamine obtained under different mini-G expression levels were consistent. Moreover, we obtained excellent dynamic ranges (Z’ factor) and the signal spans were improved for all receptor subtypes in comparison to the previously performed [35S]GTPγS binding assay.
Collapse
Affiliation(s)
- Carina Höring
- Correspondence: (C.H.); , (A.S.); Tel.: +49-941-943-4748 (C.H.); +49-941-943-4821 (A.S.)
| | | | | | | | | | | | | | | | - Andrea Strasser
- Correspondence: (C.H.); , (A.S.); Tel.: +49-941-943-4748 (C.H.); +49-941-943-4821 (A.S.)
| |
Collapse
|
18
|
Allikalt A, Laasfeld T, Ilisson M, Kopanchuk S, Rinken A. Quantitative analysis of fluorescent ligand binding to dopamine D 3 receptors using live-cell microscopy. FEBS J 2020; 288:1514-1532. [PMID: 32783364 DOI: 10.1111/febs.15519] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/14/2020] [Accepted: 07/30/2020] [Indexed: 12/29/2022]
Abstract
Dopamine receptors are G protein-coupled receptors that have several essential functions in the central nervous system. A better understanding of the regulatory mechanisms of ligand binding to the receptor may open new possibilities to affect the downstream signal transduction pathways. The majority of the available ligand binding assays use either membrane preparations, cell suspensions, or genetically modified receptors, which may give at least partially incorrect understanding of ligand binding. In this study, we implemented an assay combining fluorescence and bright-field microscopy to measure ligand binding to dopamine D3 receptors in live mammalian cells. For membrane fluorescence intensity quantification from microscopy images, we developed a machine learning-based user-friendly software membrane tools and incorporated it into a data management software aparecium that has been previously developed in our workgroup. For the experiments, a fluorescent ligand NAPS-Cy3B was synthesized by conjugating a dopaminergic antagonist N-(p-aminophenethyl)spiperone with a fluorophore Cy3B. The subnanomolar affinity of NAPS-Cy3B makes it a suitable ligand for the characterization of D3 receptors in live HEK293 cells. Using a microplate compatible automated widefield fluorescence microscope, together with the membrane tools software, enables the detection and quantification of ligand binding with a high-throughput. The live cell assay is suitable for the characterization of fluorescent ligand binding and also in the competition experiments for the screening of novel unlabeled dopaminergic ligands. We propose that this simple yet more native-like approach is feasible in GPCR research, as it enables the detection of ligand binding in an environment containing more components involved in the signal transduction cascade.
Collapse
Affiliation(s)
- Anni Allikalt
- Institute of Chemistry, University of Tartu, Estonia.,Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | | | | | | | - Ago Rinken
- Institute of Chemistry, University of Tartu, Estonia
| |
Collapse
|
19
|
Li XX, Lee JD, Massey NL, Guan C, Robertson AAB, Clark RJ, Woodruff TM. Pharmacological characterisation of small molecule C5aR1 inhibitors in human cells reveals biased activities for signalling and function. Biochem Pharmacol 2020; 180:114156. [PMID: 32682759 DOI: 10.1016/j.bcp.2020.114156] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/13/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022]
Abstract
The complement fragment C5a is a core effector of complement activation. C5a, acting through its major receptor C5aR1, exerts powerful pro-inflammatory and immunomodulatory functions. Dysregulation of the C5a-C5aR1 axis has been implicated in numerous immune disorders, and the therapeutic inhibition of this axis is therefore imperative for the treatment of these diseases. A myriad of small-molecule C5aR1 inhibitors have been developed and independently characterised over the past two decades, however the pharmacological properties of these compounds has been difficult to directly compare due to the wide discrepancies in the model, read-out, ligand dose and instrumentation implemented across individual studies. Here, we performed a systematic characterisation of the most commonly reported and clinically advanced small-molecule C5aR1 inhibitors (peptidic: PMX53, PMX205 and JPE1375; non-peptide: W545011, NDT9513727, DF2593A and CCX168). Through signalling assays measuring C5aR1-mediated cAMP and ERK1/2 signalling, and β-arrestin 2 recruitment, this study highlighted the signalling-pathway dependence of the rank order of potencies of the C5aR1 inhibitors. Functional experiments performed in primary human macrophages demonstrated the high insurmountable antagonistic potencies for the peptidic inhibitors as compared to the non-peptide compounds. Finally, wash-out studies provided novel insights into the duration of inhibition of the C5aR1 inhibitors, and confirmed the long-lasting antagonistic properties of PMX53 and CCX168. Overall, this study revealed the potent and prolonged antagonistic activities of selected peptidic C5aR1 inhibitors and the unique pharmacological profile of CCX168, which thus represent ideal candidates to fulfil diverse C5aR1 research and clinical therapeutic needs.
Collapse
Affiliation(s)
- Xaria X Li
- School of Biomedical Sciences, Australia
| | - John D Lee
- School of Biomedical Sciences, Australia
| | | | - Carolyn Guan
- The University of Queensland, St Lucia 4072, Australia; Department of Chemistry, Princeton University, Princeton, NJ 08544, United States
| | | | | | - Trent M Woodruff
- School of Biomedical Sciences, Australia; Queensland Brain Institute, Australia.
| |
Collapse
|
20
|
Ricart-Ortega M, Berizzi AE, Catena J, Malhaire F, Muñoz L, Serra C, Lebon G, Goudet C, Llebaria A. Development and validation of a mass spectrometry binding assay for mGlu5 receptor. Anal Bioanal Chem 2020; 412:5525-5535. [PMID: 32564119 DOI: 10.1007/s00216-020-02772-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/03/2020] [Accepted: 06/12/2020] [Indexed: 10/24/2022]
Abstract
Mass spectrometry (MS) binding assays are a label-free alternative to radioligand or fluorescence binding assays, so the readout is based on direct mass spectrometric detection of the test ligand. The study presented here describes the development and validation of a highly sensitive, rapid, and robust MS binding assay for the quantification of the binding of the metabotropic glutamate 5 (mGlu5) negative allosteric modulator (NAM), MPEP (2-methyl-6-phenylethynylpyridine) at the mGlu5 allosteric binding site. The LC-ESI-MS/MS (liquid chromatography-electrospray ionization-tandem mass spectrometric) analytical method was established and validated with a deuterated analogue of MPEP as an internal standard. The developed MS binding assay described here allowed for the determination of MS binding affinity estimates that were in agreement with affinity estimates obtained from a tritiated MPEP radioligand saturation binding assay, indicating the suitability of this methodology for determining affinity estimates for compounds that target mGlu5 allosteric binding sites. Graphical abstract.
Collapse
Affiliation(s)
- Maria Ricart-Ortega
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain.,IGF, CNRS, INSERM, University of Montpellier, 34094, Montpellier, France
| | - Alice E Berizzi
- IGF, CNRS, INSERM, University of Montpellier, 34094, Montpellier, France
| | - Juanlo Catena
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Fanny Malhaire
- IGF, CNRS, INSERM, University of Montpellier, 34094, Montpellier, France
| | - Lourdes Muñoz
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain.,SIMchem, Service of Synthesis of High Added Value Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Carmen Serra
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain.,SIMchem, Service of Synthesis of High Added Value Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Guillaume Lebon
- IGF, CNRS, INSERM, University of Montpellier, 34094, Montpellier, France
| | - Cyril Goudet
- IGF, CNRS, INSERM, University of Montpellier, 34094, Montpellier, France.
| | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain. .,SIMchem, Service of Synthesis of High Added Value Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain.
| |
Collapse
|
21
|
Perpiñá-Viciano C, Işbilir A, Zarca A, Caspar B, Kilpatrick LE, Hill SJ, Smit MJ, Lohse MJ, Hoffmann C. Kinetic Analysis of the Early Signaling Steps of the Human Chemokine Receptor CXCR4. Mol Pharmacol 2020; 98:72-87. [PMID: 32474443 PMCID: PMC7330677 DOI: 10.1124/mol.119.118448] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 05/06/2020] [Indexed: 01/14/2023] Open
Abstract
G protein–coupled receptors (GPCRs) are biologic switches that transduce extracellular stimuli into intracellular responses in the cell. Temporally resolving GPCR transduction pathways is key to understanding how cell signaling occurs. Here, we investigate the kinetics and dynamics of the activation and early signaling steps of the CXC chemokine receptor (CXCR) 4 in response to its natural ligands CXC chemokine ligand (CXCL) 12 and macrophage migration inhibitory factor (MIF), using Förster resonance energy transfer–based approaches. We show that CXCR4 presents a multifaceted response to CXCL12, with receptor activation (≈0.6 seconds) followed by a rearrangement in the receptor/G protein complex (≈1 seconds), a slower dimer rearrangement (≈1.7 seconds), and prolonged G protein activation (≈4 seconds). In comparison, MIF distinctly modulates every step of the transduction pathway, indicating distinct activation mechanisms and reflecting the different pharmacological properties of these two ligands. Our study also indicates that CXCR4 exhibits some degree of ligand-independent activity, a relevant feature for drug development.
Collapse
Affiliation(s)
- Cristina Perpiñá-Viciano
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Ali Işbilir
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Aurélien Zarca
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Birgit Caspar
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Laura E Kilpatrick
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Stephen J Hill
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Martine J Smit
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Martin J Lohse
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Carsten Hoffmann
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| |
Collapse
|
22
|
van der Velden WJC, Heitman LH, Rosenkilde MM. Perspective: Implications of Ligand-Receptor Binding Kinetics for Therapeutic Targeting of G Protein-Coupled Receptors. ACS Pharmacol Transl Sci 2020; 3:179-189. [PMID: 32296761 DOI: 10.1021/acsptsci.0c00012] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Indexed: 12/16/2022]
Abstract
The concept of ligand-receptor binding kinetics has been broadly applied in drug development pipelines focusing on G protein-coupled receptors (GPCRs). The ligand residence time (RT) for a receptor describes how long a ligand-receptor complex exists, and is defined as the reciprocal of the dissociation rate constant (k off). RT has turned out to be a valuable parameter for GPCR researchers focusing on drug development as a good predictor of in vivo efficacy. The positive correlation between RT and in vivo efficacy has been established for several drugs targeting class A GPCRs (e.g., the neurokinin-1 receptor (NK1R), the β2 adrenergic receptor (β2AR), and the muscarinic 3 receptor (M3R)) and for drugs targeting class B1 (e.g., the glucagon-like peptide 1 receptor (GLP-1R)). Recently, the association rate constant (k on) has gained similar attention as another parameter affecting in vivo efficacy. In the current perspective, we address the importance of studying ligand-receptor binding kinetics for therapeutic targeting of GPCRs, with an emphasis on how binding kinetics can be altered by subtle molecular changes in the ligands and/or the receptors and how such changes affect treatment outcome. Moreover, we speculate on the impact of binding kinetic parameters for functional selectivity and sustained receptor signaling from endosomal compartments; phenomena that have gained increasing interest in attempts to improve therapeutic targeting of GPCRs.
Collapse
Affiliation(s)
- Wijnand J C van der Velden
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK 2200, Denmark
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333 CC, The Netherlands
| | - Mette M Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK 2200, Denmark
| |
Collapse
|
23
|
Azalim P, do Monte FM, Rendeiro MM, Liu X, O'Doherty GA, Fontes CF, Leitão SG, Quintas LEM, Noël F. Conformational states of the pig kidney Na+/K+-ATPase differently affect bufadienolides and cardenolides: A directed structure-activity and structure-kinetics study. Biochem Pharmacol 2020; 171:113679. [DOI: 10.1016/j.bcp.2019.113679] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/21/2019] [Indexed: 10/25/2022]
|
24
|
Link R, Veiksina S, Tahk MJ, Laasfeld T, Paiste P, Kopanchuk S, Rinken A. The constitutive activity of melanocortin-4 receptors in cAMP pathway is allosterically modulated by zinc and copper ions. J Neurochem 2019; 153:346-361. [PMID: 31792980 DOI: 10.1111/jnc.14933] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 11/06/2019] [Accepted: 11/29/2019] [Indexed: 01/25/2023]
Abstract
Melanocortin-4 receptors (MC4 R) are unique among G-protein-coupled receptors (GPCRs) as they have endogenous ligands that can exhibit inverse agonistic properties in the case of elevated basal activity. It is known that the constitutive activity of GPCRs strongly affects the ligand-dependent physiological responses, but little is known about these regulatory mechanisms. Since several metal ions have been shown to be important modulators of the signal transduction of GPCRs, we hypothesized that metal ions regulate the basal activity of MC4 Rs. Implementation of a fluorescence anisotropy assay and novel redshifted fluorescent peptides enabled kinetic characterization of ligand binding to MC4 R expressed on budded baculoviruses. We show that Ca2+ is required for high-affinity ligand binding, but Zn2+ and Cu2+ in the presence of Ca2+ behave as negative allosteric modulators of ligand binding to MC4 R. FRET-based cAMP biosensor was used to measure the activation of MC4 R stably expressed in CHO-K1 cells. At low micromolar concentrations, Zn2+ caused MC4 R-dependent activation of the cAMP pathway, whereas Cu2+ reduced the activity of MC4 R even below the basal level. These findings indicate that at physiologically relevant concentrations can Zn2+ and Cu2+ function as MC4 R agonists or inverse agonists, respectively. This means that depending on the level of constitutive activity induced by Zn2+ ions, the pharmacological effect of orthosteric ligands of MC4 R can be switched from a partial to an inverse agonist. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. More information about the Open Science badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Reet Link
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Santa Veiksina
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | | | - Tõnis Laasfeld
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Päärn Paiste
- Department of Geology, University of Tartu, Tartu, Estonia
| | | | - Ago Rinken
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| |
Collapse
|
25
|
Bosma R, Wang Z, Kooistra AJ, Bushby N, Kuhne S, van den Bor J, Waring MJ, de Graaf C, de Esch IJ, Vischer HF, Sheppard RJ, Wijtmans M, Leurs R. Route to Prolonged Residence Time at the Histamine H 1 Receptor: Growing from Desloratadine to Rupatadine. J Med Chem 2019; 62:6630-6644. [PMID: 31274307 PMCID: PMC6750840 DOI: 10.1021/acs.jmedchem.9b00447] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
![]()
Drug–target
binding kinetics are an important predictor of in vivo drug efficacy,
yet the relationship
between ligand structures and their binding kinetics is often poorly
understood. We show that both rupatadine (1) and desloratadine
(2) have a long residence time at the histamine H1 receptor (H1R). Through development of a [3H]levocetirizine radiolabel, we find that the residence time
of 1 exceeds that of 2 more than 10-fold.
This was further explored with 22 synthesized rupatadine and desloratadine
analogues. Methylene-linked cycloaliphatic or β-branched substitutions
of desloratadine increase the residence time at the H1R,
conveying a longer duration of receptor antagonism. However, cycloaliphatic
substituents directly attached to the piperidine amine (i.e., lacking
the spacer) have decreased binding affinity and residence time compared
to their methylene-linked structural analogues. Guided by docking
studies, steric constraints within the binding pocket are hypothesized
to explain the observed differences in affinity and binding kinetics
between analogues.
Collapse
Affiliation(s)
- Reggie Bosma
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science , VU University Amsterdam , De Boelelaan 1083 , 1081 HV Amsterdam , The Netherlands
| | - Zhiyong Wang
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science , VU University Amsterdam , De Boelelaan 1083 , 1081 HV Amsterdam , The Netherlands
| | - Albert J Kooistra
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science , VU University Amsterdam , De Boelelaan 1083 , 1081 HV Amsterdam , The Netherlands
| | - Nick Bushby
- Operations, BioPharmaceuticals R&D , AstraZeneca , Alderley Park , Macclesfield SK10 4TG , United Kingdom
| | - Sebastiaan Kuhne
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science , VU University Amsterdam , De Boelelaan 1083 , 1081 HV Amsterdam , The Netherlands
| | - Jelle van den Bor
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science , VU University Amsterdam , De Boelelaan 1083 , 1081 HV Amsterdam , The Netherlands
| | - Michael J Waring
- Medicinal Chemistry, Research and Early Development, Oncology R&D , AstraZeneca , Alderley Park , Macclesfield SK10 4TG , United Kingdom
| | - Chris de Graaf
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science , VU University Amsterdam , De Boelelaan 1083 , 1081 HV Amsterdam , The Netherlands
| | - Iwan J de Esch
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science , VU University Amsterdam , De Boelelaan 1083 , 1081 HV Amsterdam , The Netherlands
| | - Henry F Vischer
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science , VU University Amsterdam , De Boelelaan 1083 , 1081 HV Amsterdam , The Netherlands
| | - Robert J Sheppard
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D , AstraZeneca , Gothenburg 431 50 , Sweden
| | - Maikel Wijtmans
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science , VU University Amsterdam , De Boelelaan 1083 , 1081 HV Amsterdam , The Netherlands
| | - Rob Leurs
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science , VU University Amsterdam , De Boelelaan 1083 , 1081 HV Amsterdam , The Netherlands
| |
Collapse
|
26
|
Probe dependency in the determination of ligand binding kinetics at a prototypical G protein-coupled receptor. Sci Rep 2019; 9:7906. [PMID: 31133718 PMCID: PMC6536503 DOI: 10.1038/s41598-019-44025-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/25/2019] [Indexed: 01/28/2023] Open
Abstract
Drug-target binding kinetics are suggested to be important parameters for the prediction of in vivo drug-efficacy. For G protein-coupled receptors (GPCRs), the binding kinetics of ligands are typically determined using association binding experiments in competition with radiolabelled probes, followed by analysis with the widely used competitive binding kinetics theory developed by Motulsky and Mahan. Despite this, the influence of the radioligand binding kinetics on the kinetic parameters derived for the ligands tested is often overlooked. To address this, binding rate constants for a series of histamine H1 receptor (H1R) antagonists were determined using radioligands with either slow (low koff) or fast (high koff) dissociation characteristics. A correlation was observed between the probe-specific datasets for the kinetic binding affinities, association rate constants and dissociation rate constants. However, the magnitude and accuracy of the binding rate constant-values was highly dependent on the used radioligand probe. Further analysis using recently developed fluorescent binding methods corroborates the finding that the Motulsky-Mahan methodology is limited by the employed assay conditions. The presented data suggest that kinetic parameters of GPCR ligands depend largely on the characteristics of the probe used and results should therefore be viewed within the experimental context and limitations of the applied methodology.
Collapse
|
27
|
IJzerman AP, Guo D. Drug-Target Association Kinetics in Drug Discovery. Trends Biochem Sci 2019; 44:861-871. [PMID: 31101454 DOI: 10.1016/j.tibs.2019.04.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/28/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023]
Abstract
The important role of ligand-receptor binding kinetics in drug design and discovery is increasingly recognized by the drug research community. Over the past decade, accumulating evidence has shown that optimizing the ligand's dissociation rate constant can lead to desirable duration of in vivo target occupancy and, hence, improved pharmacodynamic properties. However, the association rate constant as a pharmacological principle remains less investigated, whereas it can play an equally important role in the selection of drug candidates. This review provides a compilation and discussion of otherwise scarce and dispersed information on this topic, bringing to light the importance of drug-target association in kinetics-directed drug design and discovery.
Collapse
Affiliation(s)
- Adriaan P IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
28
|
Reiner D, Stark H. Ligand binding kinetics at histamine H3 receptors by fluorescence-polarization with real-time monitoring. Eur J Pharmacol 2019; 848:112-120. [DOI: 10.1016/j.ejphar.2019.01.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/24/2019] [Accepted: 01/24/2019] [Indexed: 01/07/2023]
|
29
|
A live cell NanoBRET binding assay allows the study of ligand-binding kinetics to the adenosine A 3 receptor. Purinergic Signal 2019; 15:139-153. [PMID: 30919204 PMCID: PMC6635573 DOI: 10.1007/s11302-019-09650-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 02/14/2019] [Indexed: 01/14/2023] Open
Abstract
There is a growing interest in understanding the binding kinetics of compounds that bind to G protein-coupled receptors prior to progressing a lead compound into clinical trials. The widely expressed adenosine A3 receptor (A3AR) has been implicated in a range of diseases including immune conditions, and compounds that aim to selectively target this receptor are currently under development for arthritis. Kinetic studies at the A3AR have been performed using a radiolabelled antagonist, but due to the kinetics of this probe, they have been carried out at 10 °C in membrane preparations. In this study, we have developed a live cell NanoBRET ligand binding assay using fluorescent A3AR antagonists to measure kinetic parameters of labelled and unlabelled compounds at the A3AR at physiological temperatures. The kinetic profiles of four fluorescent antagonists were determined in kinetic association assays, and it was found that XAC-ser-tyr-X-BY630 had the longest residence time (RT = 288 ± 62 min) at the A3AR. The association and dissociation rate constants of three antagonists PSB-11, compound 5, and LUF7565 were also determined using two fluorescent ligands (XAC-ser-tyr-X-BY630 or AV039, RT = 6.8 ± 0.8 min) as the labelled probe and compared to those obtained using a radiolabelled antagonist ([3H]PSB-11, RT = 44.6 ± 3.9 min). There was close agreement in the kinetic parameters measured with AV039 and [3H]PSB-11 but significant differences to those obtained using XAC-S-ser-S-tyr-X-BY630. These data indicate that selecting a probe with the appropriate kinetics is important to accurately determine the kinetics of unlabelled ligands with markedly different kinetic profiles.
Collapse
|
30
|
Liu Q, Herrmann A, Huang Q. Surface Binding Energy Landscapes Affect Phosphodiesterase Isoform-Specific Inhibitor Selectivity. Comput Struct Biotechnol J 2018; 17:101-109. [PMID: 30719238 PMCID: PMC6349013 DOI: 10.1016/j.csbj.2018.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/21/2018] [Accepted: 11/23/2018] [Indexed: 01/02/2023] Open
Abstract
As human phosphodiesterase (PDE) proteins are attractive drug targets, a large number of selective PDE inhibitors have been developed. However, since the catalytic sites of PDE isoforms are conserved in sequence and structure, it remains unclear how these inhibitors discriminate PDE isoforms in a selective manner. Here we perform long-time scale molecular dynamics (MD) simulations to investigate the spontaneous association processes of a highly selective PDE2A inhibitor (BAY60–7550) with the catalytic pockets of six PDE isoforms. We found that the free-energy landscapes of PDE:BAY60–7550 interactions on the PDE surfaces are very different between various PDE isoforms; and the free-energy landscape of PDE2A forms a favorable low-energy pathway that not only drives BAY60–7550 toward the target binding site, but also guides BAY60–7750 to adopt its native binding conformation known from crystal structure. Thus, this study reveals that the inhibitor interactions with the PDE surface residues play an important role in its high selectivity for PDE2A, and thereby provides new fundamental insights into the PDE isoform-specific inhibitor selectivity.
Collapse
Affiliation(s)
- Qing Liu
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Andreas Herrmann
- Institute for Biology and IRI Lifesciences, Humboldt-Universität zu Berlin, Invalidenstrasse 42, Berlin 10115, Germany
| | - Qiang Huang
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
31
|
Allikalt A, Kopanchuk S, Rinken A. Implementation of fluorescence anisotropy-based assay for the characterization of ligand binding to dopamine D1 receptors. Eur J Pharmacol 2018; 839:40-46. [DOI: 10.1016/j.ejphar.2018.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 01/11/2023]
|
32
|
Mocking TAM, Verweij EWE, Vischer HF, Leurs R. Homogeneous, Real-Time NanoBRET Binding Assays for the Histamine H 3 and H 4 Receptors on Living Cells. Mol Pharmacol 2018; 94:1371-1381. [PMID: 30249614 DOI: 10.1124/mol.118.113373] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/19/2018] [Indexed: 11/22/2022] Open
Abstract
Receptor-binding affinity and ligand-receptor residence time are key parameters for the selection of drug candidates and are routinely determined using radioligand competition-binding assays. Recently, a novel bioluminescence resonance energy transfer (BRET) method utilizing a NanoLuc-fused receptor was introduced to detect fluorescent ligand binding. Moreover, this NanoBRET method gives the opportunity to follow fluorescent ligand binding on intact cells in real time, and therefore, results might better reflect in vivo conditions as compared with the routinely used cell homogenates or purified membrane fractions. In this study, a real-time NanoBRET-based binding assay was established and validated to detect binding of unlabeled ligands to the histamine H3 receptor (H3R) and histamine H4 receptor on intact cells. Obtained residence times of clinically tested H3R antagonists were reflected by their duration of H3R antagonism in a functional receptor recovery assay.
Collapse
Affiliation(s)
- Tamara A M Mocking
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eléonore W E Verweij
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Henry F Vischer
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Rob Leurs
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Bosma R, van den Bor J, Vischer HF, Labeaga L, Leurs R. The long duration of action of the second generation antihistamine bilastine coincides with its long residence time at the histamine H 1 receptor. Eur J Pharmacol 2018; 838:107-111. [PMID: 30201377 DOI: 10.1016/j.ejphar.2018.09.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 12/15/2022]
Abstract
Drug-target binding kinetics has recently attracted considerable interest in view of the potential predictive power for in vivo drug efficacy. The recently introduced antihistamine bilastine has a long duration of in vivo drug action, which outlasts pharmacological active bilastine concentrations in blood. To provide a molecular basis for the long duration of action, we explored the kinetics of bilastine binding to the human histamine H1 receptor using [3H]mepyramine binding studies and compared its pharmacodynamics properties to the reference compounds fexofenadine and diphenhydramine, which have a long (60 ± 20 min) and short (0.41 ± 0.1 min) residence time, respectively. Bilastine shows a long drug-target residence time at the H1 receptor (73 ± 5 min) and this results in a prolonged H1 receptor antagonism in vitro (Ca2+ mobilization in Fluo-4 loaded HeLa cells), following a washout of unbound antagonist. Hence, the long residence time of bilastine can explain the observed long duration of drug action in vivo.
Collapse
Affiliation(s)
- Reggie Bosma
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Jelle van den Bor
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Henry F Vischer
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Luis Labeaga
- Clinical Research Department, Faes Farma SA, Leioa, Bizkaia, Spain
| | - Rob Leurs
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands.
| |
Collapse
|
34
|
Guo D, Peletier LA, Bridge L, Keur W, de Vries H, Zweemer A, Heitman LH, IJzerman AP. A two-state model for the kinetics of competitive radioligand binding. Br J Pharmacol 2018; 175:1719-1730. [PMID: 29486053 PMCID: PMC5913406 DOI: 10.1111/bph.14184] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 12/11/2017] [Accepted: 02/19/2018] [Indexed: 01/09/2023] Open
Abstract
Background and Purpose Ligand–receptor binding kinetics is receiving increasing attention in the drug research community. The Motulsky and Mahan model, a one‐state model, offers a method for measuring the binding kinetics of an unlabelled ligand, with the assumption that the labelled ligand has no preference while binding to distinct states or conformations of a drug target. As such, the one‐state model is not applicable if the radioligand displays biphasic binding kinetics to the receptor. Experimental Approach We extended the Motulsky and Mahan model to a two‐state model, in which the kinetics of the unlabelled competitor binding to different receptor states (R1 and R2) can be measured. With this extended model, we determined the binding kinetics of unlabelled N‐5′‐ethylcarboxamidoadenosine (NECA), a representative agonist for the adenosine A1 receptor. Subsequently, an application of the model was exemplified by measuring the binding kinetics of other A1 receptor ligands. In addition, limitations of the model were investigated as well. Key Results The kinetic rate constants of unlabelled NECA were comparable with the results of kinetic radioligand binding assays in which [3H]‐NECA was used. The model was further validated by good correlation between simulated results and the experimental data. Conclusion The two‐state model is sufficient to analyse the binding kinetics of an unlabelled ligand, when a radioligand shows biphasic association characteristics. We expect this two‐state model to have general applicability for other targets as well.
Collapse
Affiliation(s)
- Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | | | - Lloyd Bridge
- Department of Mathematics, Swansea University, Swansea, UK.,Department of Engineering Design and Mathematics, University of the West of England, Bristol, UK
| | - Wesley Keur
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Henk de Vries
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Annelien Zweemer
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Laura H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| |
Collapse
|
35
|
Hoare SRJ, Pierre N, Moya AG, Larson B. Kinetic operational models of agonism for G-protein-coupled receptors. J Theor Biol 2018; 446:168-204. [PMID: 29486201 DOI: 10.1016/j.jtbi.2018.02.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 02/07/2018] [Accepted: 02/13/2018] [Indexed: 01/06/2023]
Abstract
The application of kinetics to research and therapeutic development of G-protein-coupled receptors has become increasingly valuable. Pharmacological models provide the foundation of pharmacology, providing concepts and measurable parameters such as efficacy and potency that have underlain decades of successful drug discovery. Currently there are few pharmacological models that incorporate kinetic activity in such a way as to yield experimentally-accessible drug parameters. In this study, a kinetic model of pharmacological response was developed that provides a kinetic descriptor of efficacy (the transduction rate constant, kτ) and allows measurement of receptor-ligand binding kinetics from functional data. The model assumes: (1) receptor interacts with a precursor of the response ("Transduction potential") and converts it to the response. (2) The response can decay. Familiar response vs time plots emerge, depending on whether transduction potential is depleted and/or response decays. These are the straight line, the "association" exponential curve, and the rise-and-fall curve. Convenient, familiar methods are described for measuring the model parameters and files are provided for the curve-fitting program Prism (GraphPad Software) that can be used as a guide. The efficacy parameter kτ is straightforward to measure and accounts for receptor reserve; all that is required is measurement of response over time at a maximally-stimulating concentration of agonist. The modular nature of the model framework allows it to be extended. Here this is done to incorporate antagonist-receptor binding kinetics and slow agonist-receptor equilibration. In principle, the modular framework can incorporate other cellular processes, such as receptor desensitization. The kinetic response model described here can be applied to measure kinetic pharmacological parameters than can be used to advance the understanding of GPCR pharmacology and optimize new and improved therapeutics.
Collapse
Affiliation(s)
- Samuel R J Hoare
- Pharmechanics, LLC, 14 Sunnyside Drive South, Owego NY 13827, USA.
| | | | | | - Brad Larson
- BioTek Instruments, Inc, 100 Tigan Street, Winooski, VT 05404, USA
| |
Collapse
|
36
|
Assays with Detection of Fluorescence Anisotropy: Challenges and Possibilities for Characterizing Ligand Binding to GPCRs. Trends Pharmacol Sci 2018; 39:187-199. [DOI: 10.1016/j.tips.2017.10.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/10/2017] [Accepted: 10/10/2017] [Indexed: 01/24/2023]
|
37
|
Stoddart LA, Vernall AJ, Bouzo-Lorenzo M, Bosma R, Kooistra AJ, de Graaf C, Vischer HF, Leurs R, Briddon SJ, Kellam B, Hill SJ. Development of novel fluorescent histamine H 1-receptor antagonists to study ligand-binding kinetics in living cells. Sci Rep 2018; 8:1572. [PMID: 29371669 PMCID: PMC5785503 DOI: 10.1038/s41598-018-19714-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/03/2018] [Indexed: 01/01/2023] Open
Abstract
The histamine H1-receptor (H1R) is an important mediator of allergy and inflammation. H1R antagonists have particular clinical utility in allergic rhinitis and urticaria. Here we have developed six novel fluorescent probes for this receptor that are very effective for high resolution confocal imaging, alongside bioluminescence resonance energy transfer approaches to monitor H1R ligand binding kinetics in living cells. The latter technology exploits the opportunities provided by the recently described bright bioluminescent protein NanoLuc when it is fused to the N-terminus of a receptor. Two different pharmacophores (mepyramine or the fragment VUF13816) were used to generate fluorescent H1R antagonists conjugated via peptide linkers to the fluorophore BODIPY630/650. Kinetic properties of the probes showed wide variation, with the VUF13816 analogues having much longer H1R residence times relative to their mepyramine-based counterparts. The kinetics of these fluorescent ligands could also be monitored in membrane preparations providing new opportunities for future drug discovery applications.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Andrea J Vernall
- School of Pharmacy, Division of Biomolecular Science and Medicinal Chemistry, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Monica Bouzo-Lorenzo
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Reggie Bosma
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Albert J Kooistra
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Chris de Graaf
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Henry F Vischer
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Rob Leurs
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Stephen J Briddon
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Barrie Kellam
- School of Pharmacy, Division of Biomolecular Science and Medicinal Chemistry, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK.
| | - Stephen J Hill
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK.
| |
Collapse
|
38
|
Guo D, IJzerman AP. Molecular Basis of Ligand Dissociation from G Protein-Coupled Receptors and Predicting Residence Time. Methods Mol Biol 2018; 1705:197-206. [PMID: 29188564 DOI: 10.1007/978-1-4939-7465-8_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
G protein-coupled receptors (GPCRs) are integral membrane proteins and represent the largest class of drug targets. During the past decades progress in structural biology has enabled the crystallographic elucidation of the architecture of these important macromolecules. It also provided atomic-level visualization of ligand-receptor interactions, dramatically boosting the impact of structure-based approaches in drug discovery. However, knowledge obtained through crystallography is limited to static structural information. Less information is available showing how a ligand associates with or dissociates from a given receptor, whose importance is in fact increasingly recognized by the drug research community. Owing to recent advances in computer power and algorithms, molecular dynamics stimulations have become feasible that help in analyzing the kinetics of the ligand binding process. Here, we review what is currently known about the dynamics of GPCRs in the context of ligand association and dissociation, as determined through both crystallography and computer simulations. We particularly focus on the molecular basis of ligand dissociation from GPCRs and provide case studies that predict ligand dissociation pathways and residence time.
Collapse
Affiliation(s)
- Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands.
| |
Collapse
|
39
|
Bosma R, Witt G, Vaas LAI, Josimovic I, Gribbon P, Vischer HF, Gul S, Leurs R. The Target Residence Time of Antihistamines Determines Their Antagonism of the G Protein-Coupled Histamine H1 Receptor. Front Pharmacol 2017; 8:667. [PMID: 29033838 PMCID: PMC5627017 DOI: 10.3389/fphar.2017.00667] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/07/2017] [Indexed: 11/13/2022] Open
Abstract
The pharmacodynamics of drug-candidates is often optimized by metrics that describe target binding (Kd or Ki value) or target modulation (IC50). However, these metrics are determined at equilibrium conditions, and consequently information regarding the onset and offset of target engagement and modulation is lost. Drug-target residence time is a measure for the lifetime of the drug-target complex, which has recently been receiving considerable interest, as target residence time is shown to have prognostic value for the in vivo efficacy of several drugs. In this study, we have investigated the relation between the increased residence time of antihistamines at the histamine H1 receptor (H1R) and the duration of effective target-inhibition by these antagonists. Hela cells, endogenously expressing low levels of the H1R, were incubated with a series of antihistamines and dissociation was initiated by washing away the unbound antihistamines. Using a calcium-sensitive fluorescent dye and a label free, dynamic mass redistribution based assay, functional recovery of the H1R responsiveness was measured by stimulating the cells with histamine over time, and the recovery was quantified as the receptor recovery time. Using these assays, we determined that the receptor recovery time for a set of antihistamines differed more than 40-fold and was highly correlated to their H1R residence times, as determined with competitive radioligand binding experiments to the H1R in a cell homogenate. Thus, the receptor recovery time is proposed as a cell-based and physiologically relevant metric for the lead optimization of G protein-coupled receptor antagonists, like the H1R antagonists. Both, label-free or real-time, classical signaling assays allow an efficient and physiologically relevant determination of kinetic properties of drug molecules.
Collapse
Affiliation(s)
- Reggie Bosma
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Lea A I Vaas
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Ivana Josimovic
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Philip Gribbon
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Henry F Vischer
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Rob Leurs
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| |
Collapse
|
40
|
Allikalt A, Rinken A. Budded baculovirus particles as a source of membrane proteins for radioligand binding assay: The case of dopamine D 1 receptor. J Pharmacol Toxicol Methods 2017; 86:81-86. [DOI: 10.1016/j.vascn.2017.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/16/2017] [Accepted: 04/08/2017] [Indexed: 01/27/2023]
|
41
|
Vanderheyden PML, Benachour N. Influence of the cellular environment on ligand binding kinetics at membrane-bound targets. Bioorg Med Chem Lett 2017; 27:3621-3628. [PMID: 28666735 DOI: 10.1016/j.bmcl.2017.06.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 10/19/2022]
Abstract
While historically 'in vitro' binding data were obtained by analyzing equilibrium experiments, kinetic data are increasingly appreciated to provide information on the time a particular compound remains bound to its target. This information is of biological importance to understand the molecular mechanism of a drug not only to evaluate the time a particular receptor/enzyme is blocked in the case of antagonists/inhibitors but also to investigate its contribution to the efficacy to mediate signaling in the case of agonists. There is accumulating evidence that many drugs binding to either membrane-bound receptors or enzymes are found to display long duration of action which can be ascribed to slow dissociation from their target proteins. In the present review three such examples are discussed which encompass ligands that bind to membrane-bound proteins and from which it appears that the tight binding kinetics is influenced by the cellular/membrane environment of the target protein.
Collapse
Affiliation(s)
- Patrick M L Vanderheyden
- Research Group of Molecular and Biochemical Pharmacology, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, VUB-MBFA, Pleinlaan 2, B-1050 Brussels, Belgium.
| | - Nerdjes Benachour
- Research Group of Molecular and Biochemical Pharmacology, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, VUB-MBFA, Pleinlaan 2, B-1050 Brussels, Belgium
| |
Collapse
|
42
|
Gendron L, Cahill CM, von Zastrow M, Schiller PW, Pineyro G. Molecular Pharmacology of δ-Opioid Receptors. Pharmacol Rev 2017; 68:631-700. [PMID: 27343248 DOI: 10.1124/pr.114.008979] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Opioids are among the most effective analgesics available and are the first choice in the treatment of acute severe pain. However, partial efficacy, a tendency to produce tolerance, and a host of ill-tolerated side effects make clinically available opioids less effective in the management of chronic pain syndromes. Given that most therapeutic opioids produce their actions via µ-opioid receptors (MOPrs), other targets are constantly being explored, among which δ-opioid receptors (DOPrs) are being increasingly considered as promising alternatives. This review addresses DOPrs from the perspective of cellular and molecular determinants of their pharmacological diversity. Thus, DOPr ligands are examined in terms of structural and functional variety, DOPrs' capacity to engage a multiplicity of canonical and noncanonical G protein-dependent responses is surveyed, and evidence supporting ligand-specific signaling and regulation is analyzed. Pharmacological DOPr subtypes are examined in light of the ability of DOPr to organize into multimeric arrays and to adopt multiple active conformations as well as differences in ligand kinetics. Current knowledge on DOPr targeting to the membrane is examined as a means of understanding how these receptors are especially active in chronic pain management. Insight into cellular and molecular mechanisms of pharmacological diversity should guide the rational design of more effective, longer-lasting, and better-tolerated opioid analgesics for chronic pain management.
Collapse
Affiliation(s)
- Louis Gendron
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Catherine M Cahill
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Mark von Zastrow
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Peter W Schiller
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Graciela Pineyro
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| |
Collapse
|
43
|
Estimation of kinetic and thermodynamic ligand-binding parameters using computational strategies. Future Med Chem 2017; 9:507-523. [DOI: 10.4155/fmc-2016-0224] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Kinetic and thermodynamic ligand–protein binding parameters are gaining growing importance as key information to consider in drug discovery. The determination of the molecular structures, using particularly x-ray and NMR techniques, is crucial for understanding how a ligand recognizes its target in the final binding complex. However, for a better understanding of the recognition processes, experimental studies of ligand–protein interactions are needed. Even though several techniques can be used to investigate both thermodynamic and kinetic profiles for a ligand–protein complex, these procedures are very often laborious, time consuming and expensive. In the last 10 years, computational approaches have enormous potential in providing insights into each of the above effects and in parsing their contributions to the changes in both kinetic and thermodynamic binding parameters. The main purpose of this review is to summarize the state of the art of computational strategies for estimating the kinetic and thermodynamic parameters of a ligand–protein binding.
Collapse
|
44
|
Riddy DM, Cook AE, Diepenhorst NA, Bosnyak S, Brady R, Mannoury la Cour C, Mocaer E, Summers RJ, Charman WN, Sexton PM, Christopoulos A, Langmead CJ. Isoform-Specific Biased Agonism of Histamine H3 Receptor Agonists. Mol Pharmacol 2016; 91:87-99. [PMID: 27864425 DOI: 10.1124/mol.116.106153] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 11/17/2016] [Indexed: 12/11/2022] Open
Abstract
The human histamine H3 receptor (hH3R) is subject to extensive gene splicing that gives rise to a large number of functional and nonfunctional isoforms. Despite the general acceptance that G protein-coupled receptors can adopt different ligand-induced conformations that give rise to biased signaling, this has not been studied for the H3R; further, it is unknown whether splice variants of the same receptor engender the same or differential biased signaling. Herein, we profiled the pharmacology of histamine receptor agonists at the two most abundant hH3R splice variants (hH3R445 and hH3R365) across seven signaling endpoints. Both isoforms engender biased signaling, notably for 4-[3-(benzyloxy)propyl]-1H-imidazole (proxyfan) [e.g., strong bias toward phosphorylation of glycogen synthase kinase 3β (GSK3β) via the full-length receptor] and its congener 3-(1H-imidazol-4-yl)propyl-(4-iodophenyl)-methyl ether (iodoproxyfan), which are strongly consistent with the former's designation as a "protean" agonist. The 80 amino acid IL3 deleted isoform hH3R365 is more permissive in its signaling than hH3R445: 2-(1H-imidazol-5-yl)ethyl imidothiocarbamate (imetit), proxyfan, and iodoproxyfan were all markedly biased away from calcium signaling, and principal component analysis of the full data set revealed divergent profiles for all five agonists. However, most interesting was the identification of differential biased signaling between the two isoforms. Strikingly, hH3R365 was completely unable to stimulate GSK3β phosphorylation, an endpoint robustly activated by the full-length receptor. To the best of our knowledge, this is the first quantitative example of differential biased signaling via isoforms of the same G protein-coupled receptor that are simultaneously expressed in vivo and gives rise to the possibility of selective pharmacological targeting of individual receptor splice variants.
Collapse
Affiliation(s)
- Darren M Riddy
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - Anna E Cook
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - Natalie A Diepenhorst
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - Sanja Bosnyak
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - Ryan Brady
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - Clotilde Mannoury la Cour
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - Elisabeth Mocaer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - William N Charman
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| | - Christopher J Langmead
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., A.E.C., N.A.D., S.B., R.B., R.J.S., W.N.C., P.M.S., A.C., C.J.L.); and Institut de Recherches Internationales Servier, Suresnes, France (C.M.C., E.M.)
| |
Collapse
|
45
|
Dynamics of ligand binding to GPCR: Residence time of melanocortins and its modulation. Pharmacol Res 2016; 113:747-753. [DOI: 10.1016/j.phrs.2016.05.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/22/2016] [Accepted: 05/27/2016] [Indexed: 01/06/2023]
|
46
|
A new, simple and robust radioligand binding method used to determine kinetic off-rate constants for unlabeled ligands. Application at α 2A - and α 2C -adrenoceptors. Eur J Pharmacol 2016; 788:113-121. [DOI: 10.1016/j.ejphar.2016.06.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/12/2016] [Accepted: 06/15/2016] [Indexed: 10/21/2022]
|
47
|
Heifetz A, James T, Morao I, Bodkin MJ, Biggin PC. Guiding lead optimization with GPCR structure modeling and molecular dynamics. Curr Opin Pharmacol 2016; 30:14-21. [DOI: 10.1016/j.coph.2016.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/16/2016] [Accepted: 06/17/2016] [Indexed: 01/04/2023]
|
48
|
BRET-based β-arrestin2 recruitment to the histamine H1 receptor for investigating antihistamine binding kinetics. Pharmacol Res 2016; 111:679-687. [PMID: 27468652 DOI: 10.1016/j.phrs.2016.07.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/14/2016] [Accepted: 07/24/2016] [Indexed: 01/23/2023]
Abstract
Ligand residence time is thought to be a critical parameter for optimizing the in vivo efficacy of drug candidates. For the histamine H1 receptor (H1R) and other G protein-coupled receptors, the kinetics of ligand binding are typically measured by low throughput radioligand binding experiments using homogenized cell membranes expressing the target receptor. In this study, a real-time proximity assay between H1R and β-arrestin2 in living cells was established to investigate the dynamics of antihistamine binding to the H1R. No receptor reserve was found for the histamine-induced recruitment of β-arrestin2 to the H1R and the transiently recruited β-arrestin2 therefore reflected occupancy of the receptor by histamine. Antihistamines displayed similar kinetic signatures on antagonizing histamine-induced β-arrestin2 recruitment as compared to displacing radioligand binding from the H1R. This homogeneous functional method unambiguously determined the fifty-fold difference in the dissociation rate constant between mepyramine and the long residence time antihistamines levocetirizine and desloratadine.
Collapse
|
49
|
Impact, determination and prediction of drug-receptor residence times for GPCRs. Curr Opin Pharmacol 2016; 30:22-26. [PMID: 27428776 DOI: 10.1016/j.coph.2016.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/03/2016] [Accepted: 07/04/2016] [Indexed: 01/02/2023]
Abstract
The residence time of a ligand on a GPCR of interest has become an optimization parameter in many examples in drug design. Long residence times can counterbalance unfavorable pharmacokinetic parameters, contributing to compound safety, and short residence times can be a tool to avoid target related side effects. Unlike the prediction and interpretation of the structure-activity relationship (SAR) of a ligand class on a receptor, the understanding and prediction of the structure-kinetics relationship (SKR) is much more demanding. Experimental and computational approaches are described, which serve to either rationalize SKR or to predict the kinetic parameters such as on-rates and off-rates.
Collapse
|
50
|
Guo D, Heitman LH, IJzerman AP. Kinetic Aspects of the Interaction between Ligand and G Protein-Coupled Receptor: The Case of the Adenosine Receptors. Chem Rev 2016; 117:38-66. [DOI: 10.1021/acs.chemrev.6b00025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Dong Guo
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Laura H. Heitman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| |
Collapse
|