1
|
Patera F, Mistry SJ, Kindon ND, Comeo E, Goulding J, Kellam B, Kilpatrick LE, Franks H, Hill SJ. A novel and selective fluorescent ligand for the study of adenosine A 2B receptors. Pharmacol Res Perspect 2024; 12:e1223. [PMID: 39031734 PMCID: PMC11191602 DOI: 10.1002/prp2.1223] [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: 02/16/2024] [Revised: 04/25/2024] [Accepted: 05/27/2024] [Indexed: 07/22/2024] Open
Abstract
Fluorescent ligands have proved to be powerful tools in the study of G protein-coupled receptors in living cells. Here we have characterized a new fluorescent ligand PSB603-BY630 that has high selectivity for the human adenosine A2B receptor (A2BR). The A2BR appears to play an important role in regulating immune responses in the tumor microenvironment. Here we have used PSB603-BY630 to monitor specific binding to A2BRs in M1- and M2-like macrophages derived from CD14+ human monocytes. PSB603-BY630 bound with high affinity (18.3 nM) to nanoluciferase-tagged A2BRs stably expressed in HEK293G cells. The ligand exhibited very high selectivity for the A2BR with negligible specific-binding detected at NLuc-A2AR, NLuc-A1R, or NLuc-A3R receptors at concentrations up to 500 nM. Competition binding studies showed the expected pharmacology at A2BR with the A2BR-selective ligands PSB603 and MRS-1706 demonstrating potent inhibition of the specific binding of 50 nM PSB603-BY630 to A2BR. Functional studies in HEK293G cells using Glosensor to monitor Gs-coupled cyclic AMP responses indicated that PSB603-BY630 acted as a negative allosteric regular of the agonist responses to BAY 60-6583. Furthermore, flow cytometry analysis confirmed that PSB603-BY630 could be used to selectively label endogenous A2BRs expressed on human macrophages. This ligand should be an important addition to the library of fluorescent ligands which are selective for the different adenosine receptor subtypes, and will enable study of the role of A2BRs on immune cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Foteini Patera
- Division of Physiology, Pharmacology and Neuroscience, School of Life SciencesUniversity of NottinghamNottinghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
- Centre for Cancer Sciences, School of Medicine, Biodiscovery InstituteUniversity of NottinghamNottinghamUK
| | - Sarah J. Mistry
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
- Division of Biomolecular Science and Medicinal Chemistry, School of Pharmacy, Biodiscovery InstituteUniversity of NottinghamNottinghamUK
| | - Nicholas D. Kindon
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
- Division of Biomolecular Science and Medicinal Chemistry, School of Pharmacy, Biodiscovery InstituteUniversity of NottinghamNottinghamUK
| | - Eleonora Comeo
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
- Division of Biomolecular Science and Medicinal Chemistry, School of Pharmacy, Biodiscovery InstituteUniversity of NottinghamNottinghamUK
| | - Joelle Goulding
- Division of Physiology, Pharmacology and Neuroscience, School of Life SciencesUniversity of NottinghamNottinghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
| | - Barrie Kellam
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
- Division of Biomolecular Science and Medicinal Chemistry, School of Pharmacy, Biodiscovery InstituteUniversity of NottinghamNottinghamUK
| | - Laura E. Kilpatrick
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
- Division of Biomolecular Science and Medicinal Chemistry, School of Pharmacy, Biodiscovery InstituteUniversity of NottinghamNottinghamUK
| | - Hester Franks
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
- Centre for Cancer Sciences, School of Medicine, Biodiscovery InstituteUniversity of NottinghamNottinghamUK
- Department of OncologyNottingham University Hospitals NHS TrustUK
| | - Stephen J. Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life SciencesUniversity of NottinghamNottinghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
| |
Collapse
|
2
|
Knight R, Kilpatrick LE, Hill SJ, Stocks MJ. Design, Synthesis, and Evaluation of a New Chemotype Fluorescent Ligand for the P2Y 2 Receptor. ACS Med Chem Lett 2024; 15:1127-1135. [PMID: 39015271 PMCID: PMC11247638 DOI: 10.1021/acsmedchemlett.4c00211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
The P2Y2 receptor (P2Y2R) is a target for diseases including cancer, idiopathic pulmonary fibrosis, and atherosclerosis. However, there are insufficient P2Y2R antagonists available for validating P2Y2R function and future drug development. Evaluation of how (R)-5-(7-chloro-2-((2-ethoxyethyl)amino)-4H-benzo[5,6]cyclohepta[1,2-d]thiazol-4-yl)-1-methyl-4-thioxo-3,4-dihydropyrimidin-2(1H)-one, a previously published thiazole-based analogue of AR-C118925, binds in a P2Y2R homology model was used to design new P2Y2R antagonist scaffolds. One P2Y2R antagonist scaffold retained millimolar affinity for the P2Y2R and upon further functionalization with terminal carboxylic acid groups affinity was improved over 100-fold. This functionalized P2Y2R antagonist scaffold was employed to develop new chemotype P2Y2R fluorescent ligands, that were attainable in a convergent five-step synthesis. One of these fluorescent ligands demonstrated micromolar affinity (pK d = 6.02 ± 0.12, n = 5) for the P2Y2R in isolated cell membranes and distinct pharmacology from an existing P2Y2R fluorescent antagonist, suggesting it may occupy a different binding site on the P2Y2R.
Collapse
Affiliation(s)
- Rebecca Knight
- Division
of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
- Centre
of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands NG7 2UH, U.K.
| | - Laura E. Kilpatrick
- Division
of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
- Centre
of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands NG7 2UH, U.K.
| | - Stephen J. Hill
- Centre
of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands NG7 2UH, U.K.
- Division
of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, U.K.
| | - Michael J. Stocks
- Division
of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
| |
Collapse
|
3
|
Beerkens BL, Snijders IM, Snoeck J, Liu R, Tool ATJ, Le Dévédec SE, Jespers W, Kuijpers TW, van Westen GJ, Heitman LH, IJzerman AP, van der Es D. Development of an Affinity-Based Probe to Profile Endogenous Human Adenosine A3 Receptor Expression. J Med Chem 2023; 66:11399-11413. [PMID: 37531576 PMCID: PMC10461224 DOI: 10.1021/acs.jmedchem.3c00854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Indexed: 08/04/2023]
Abstract
The adenosine A3 receptor (A3AR) is a G protein-coupled receptor (GPCR) that exerts immunomodulatory effects in pathophysiological conditions such as inflammation and cancer. Thus far, studies toward the downstream effects of A3AR activation have yielded contradictory results, thereby motivating the need for further investigations. Various chemical and biological tools have been developed for this purpose, ranging from fluorescent ligands to antibodies. Nevertheless, these probes are limited by their reversible mode of binding, relatively large size, and often low specificity. Therefore, in this work, we have developed a clickable and covalent affinity-based probe (AfBP) to target the human A3AR. Herein, we show validation of the synthesized AfBP in radioligand displacement, SDS-PAGE, and confocal microscopy experiments as well as utilization of the AfBP for the detection of endogenous A3AR expression in flow cytometry experiments. Ultimately, this AfBP will aid future studies toward the expression and function of the A3AR in pathologies.
Collapse
Affiliation(s)
- Bert L.
H. Beerkens
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Inge M. Snijders
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Joep Snoeck
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Rongfang Liu
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Anton T. J. Tool
- Department
of Molecular Hematology, Sanquin Research, Plesmalaan 125, 1066 CX Amsterdam, The Netherlands
| | - Sylvia E. Le Dévédec
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Willem Jespers
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Taco W. Kuijpers
- Department
of Molecular Hematology, Sanquin Research, Plesmalaan 125, 1066 CX Amsterdam, The Netherlands
- Department
of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma
Children’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Gerard J.P. van Westen
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Laura H. Heitman
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
- Oncode
Institute, Einsteinweg
55, 2333 CC Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Daan van der Es
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| |
Collapse
|
4
|
The chronological evolution of fluorescent GPCR probes for bioimaging. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
5
|
Wall MJ, Hill E, Huckstepp R, Barkan K, Deganutti G, Leuenberger M, Preti B, Winfield I, Carvalho S, Suchankova A, Wei H, Safitri D, Huang X, Imlach W, La Mache C, Dean E, Hume C, Hayward S, Oliver J, Zhao FY, Spanswick D, Reynolds CA, Lochner M, Ladds G, Frenguelli BG. Selective activation of Gαob by an adenosine A 1 receptor agonist elicits analgesia without cardiorespiratory depression. Nat Commun 2022; 13:4150. [PMID: 35851064 PMCID: PMC9293909 DOI: 10.1038/s41467-022-31652-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 06/23/2022] [Indexed: 02/06/2023] Open
Abstract
The development of therapeutic agonists for G protein-coupled receptors (GPCRs) is hampered by the propensity of GPCRs to couple to multiple intracellular signalling pathways. This promiscuous coupling leads to numerous downstream cellular effects, some of which are therapeutically undesirable. This is especially the case for adenosine A1 receptors (A1Rs) whose clinical potential is undermined by the sedation and cardiorespiratory depression caused by conventional agonists. We have discovered that the A1R-selective agonist, benzyloxy-cyclopentyladenosine (BnOCPA), is a potent and powerful analgesic but does not cause sedation, bradycardia, hypotension or respiratory depression. This unprecedented discrimination between native A1Rs arises from BnOCPA's unique and exquisitely selective activation of Gob among the six Gαi/o subtypes, and in the absence of β-arrestin recruitment. BnOCPA thus demonstrates a highly-specific Gα-selective activation of the native A1R, sheds new light on GPCR signalling, and reveals new possibilities for the development of novel therapeutics based on the far-reaching concept of selective Gα agonism.
Collapse
Affiliation(s)
- Mark J Wall
- School of Life Sciences, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK.
| | - Emily Hill
- School of Life Sciences, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK
| | - Robert Huckstepp
- School of Life Sciences, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK
| | - Kerry Barkan
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Giuseppe Deganutti
- Centre for Sport, Exercise and Life Sciences (CSELS), Faculty of Health and Life Sciences, Coventry University, Coventry, CV1 2DS, UK
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Michele Leuenberger
- Institute of Biochemistry and Molecular Medicine, University of Bern, 3012, Bern, Switzerland
| | - Barbara Preti
- Institute of Biochemistry and Molecular Medicine, University of Bern, 3012, Bern, Switzerland
| | - Ian Winfield
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Sabrina Carvalho
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Anna Suchankova
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | | | - Dewi Safitri
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
- Pharmacology and Clinical Pharmacy Research Group, School of Pharmacy, Bandung Institute of Technology, Bandung, 40132, Indonesia
| | - Xianglin Huang
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Wendy Imlach
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Innovation Walk, Clayton, VIC, 3800, Australia
| | - Circe La Mache
- School of Life Sciences, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK
| | - Eve Dean
- School of Life Sciences, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK
| | - Cherise Hume
- School of Life Sciences, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK
| | - Stephanie Hayward
- School of Life Sciences, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK
| | - Jess Oliver
- School of Life Sciences, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK
| | | | - David Spanswick
- NeuroSolutions Ltd, Coventry, UK
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Innovation Walk, Clayton, VIC, 3800, Australia
- Warwick Medical School, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK
| | - Christopher A Reynolds
- Centre for Sport, Exercise and Life Sciences (CSELS), Faculty of Health and Life Sciences, Coventry University, Coventry, CV1 2DS, UK
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Martin Lochner
- Institute of Biochemistry and Molecular Medicine, University of Bern, 3012, Bern, Switzerland
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK.
| | - Bruno G Frenguelli
- School of Life Sciences, University of Warwick, Gibbet Hill Rd, Coventry, CV4 7AL, UK.
| |
Collapse
|
6
|
Kok ZY, Stoddart LA, Mistry SJ, Mocking TAM, Vischer HF, Leurs R, Hill SJ, Mistry SN, Kellam B. Optimization of Peptide Linker-Based Fluorescent Ligands for the Histamine H 1 Receptor. J Med Chem 2022; 65:8258-8288. [PMID: 35734860 PMCID: PMC9234962 DOI: 10.1021/acs.jmedchem.2c00125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The histamine H1 receptor (H1R) has recently been implicated in mediating cell proliferation and cancer progression; therefore, high-affinity H1R-selective fluorescent ligands are desirable tools for further investigation of this behavior in vitro and in vivo. We previously reported a H1R fluorescent ligand, bearing a peptide-linker, based on antagonist VUF13816 and sought to further explore structure-activity relationships (SARs) around the linker, orthostere, and fluorescent moieties. Here, we report a series of high-affinity H1R fluorescent ligands varying in peptide linker composition, orthosteric targeting moiety, and fluorophore. Incorporation of a boron-dipyrromethene (BODIPY) 630/650-based fluorophore conferred high binding affinity to our H1R fluorescent ligands, remarkably overriding the linker SAR observed in corresponding unlabeled congeners. Compound 31a, both potent and subtype-selective, enabled H1R visualization using confocal microscopy at a concentration of 10 nM. Molecular docking of 31a with the human H1R predicts that the optimized peptide linker makes interactions with key residues in the receptor.
Collapse
Affiliation(s)
- Zhi Yuan Kok
- Division of Biomolecular Science and Medicinal Chemistry, School of Pharmacy, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, U.K.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, the Midlands, Nottingham NG7 2UH, U.K
| | - Leigh A Stoddart
- Division of Physiology, Pharmacology & Neuroscience, Medical School, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, U.K.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, the Midlands, Nottingham NG7 2UH, U.K
| | - Sarah J Mistry
- Division of Biomolecular Science and Medicinal Chemistry, School of Pharmacy, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, U.K.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, the Midlands, Nottingham NG7 2UH, U.K
| | - Tamara A M Mocking
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelean 1083, 1083 HV Amsterdam, The Netherlands
| | - Henry F Vischer
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelean 1083, 1083 HV Amsterdam, The Netherlands
| | - Rob Leurs
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelean 1083, 1083 HV Amsterdam, The Netherlands
| | - Stephen J Hill
- Division of Physiology, Pharmacology & Neuroscience, Medical School, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, U.K.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, the Midlands, Nottingham NG7 2UH, U.K
| | - Shailesh N Mistry
- Division of Biomolecular Science and Medicinal Chemistry, School of Pharmacy, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, U.K
| | - Barrie Kellam
- Division of Biomolecular Science and Medicinal Chemistry, School of Pharmacy, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, U.K.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, the Midlands, Nottingham NG7 2UH, U.K
| |
Collapse
|
7
|
Borgarelli C, Klingl YE, Escamilla-Ayala A, Munck S, Van Den Bosch L, De Borggraeve WM, Ismalaj E. Lighting Up the Plasma Membrane: Development and Applications of Fluorescent Ligands for Transmembrane Proteins. Chemistry 2021; 27:8605-8641. [PMID: 33733502 DOI: 10.1002/chem.202100296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Indexed: 12/16/2022]
Abstract
Despite the fact that transmembrane proteins represent the main therapeutic targets for decades, complete and in-depth knowledge about their biochemical and pharmacological profiling is not fully available. In this regard, target-tailored small-molecule fluorescent ligands are a viable approach to fill in the missing pieces of the puzzle. Such tools, coupled with the ability of high-precision optical techniques to image with an unprecedented resolution at a single-molecule level, helped unraveling many of the conundrums related to plasma proteins' life-cycle and druggability. Herein, we review the recent progress made during the last two decades in fluorescent ligand design and potential applications in fluorescence microscopy of voltage-gated ion channels, ligand-gated ion channels and G-coupled protein receptors.
Collapse
Affiliation(s)
- Carlotta Borgarelli
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven Campus Arenberg Celestijnenlaan 200F -, box 2404, 3001, Leuven, Belgium
| | - Yvonne E Klingl
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Neurobiology, VIB, Center for Brain &, Disease Research, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Abril Escamilla-Ayala
- Center for Brain & Disease Research, & VIB BioImaging Core, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Department of Neurosciences, Leuven Brain Institute, KU Leuven, Campus Gasthuisberg O&N5 - box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Sebastian Munck
- Center for Brain & Disease Research, & VIB BioImaging Core, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Department of Neurosciences, Leuven Brain Institute, KU Leuven, Campus Gasthuisberg O&N5 - box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Neurobiology, VIB, Center for Brain &, Disease Research, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Wim M De Borggraeve
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven Campus Arenberg Celestijnenlaan 200F -, box 2404, 3001, Leuven, Belgium
| | - Ermal Ismalaj
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven Campus Arenberg Celestijnenlaan 200F -, box 2404, 3001, Leuven, Belgium
| |
Collapse
|
8
|
Goulding J, Mistry SJ, Soave M, Woolard J, Briddon SJ, White CW, Kellam B, Hill SJ. Subtype selective fluorescent ligands based on ICI 118,551 to study the human β2-adrenoceptor in CRISPR/Cas9 genome-edited HEK293T cells at low expression levels. Pharmacol Res Perspect 2021; 9:e00779. [PMID: 34003582 PMCID: PMC8130569 DOI: 10.1002/prp2.779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022] Open
Abstract
Fluorescent ligand technologies have proved to be powerful tools to improve our understanding of ligand‐receptor interactions. Here we have characterized a small focused library of nine fluorescent ligands based on the highly selective β2‐adrenoceptor (β2AR) antagonist ICI 118,551. The majority of fluorescent ICI 118,551 analogs had good affinity for the β2AR (pKD >7.0) with good selectivity over the β1AR (pKD <6.0). The most potent and selective ligands being 8c (ICI 118,551‐Gly‐Ala‐BODIPY‐FL‐X; β2AR pKD 7.48), 9c (ICI 118,551‐βAla‐βAla‐BODIPY‐FL‐X; β2AR pKD 7.48), 12a (ICI 118,551‐PEG‐BODIPY‐X‐630/650; β2AR pKD 7.56), and 12b (ICI 118,551‐PEG‐BODIPY‐FL; β2AR pKD 7.42). 9a (ICI 118,551‐βAla‐βAla‐BODIPY‐X‐630/650) had the highest affinity at recombinant β2ARs (pKD 7.57), but also exhibited significant binding affinity to the β1AR (pKD 6.69). Nevertheless, among the red fluorescent ligands, 9a had the best imaging characteristics in recombinant HEK293 T cells and labeling was mostly confined to the cell surface. In contrast, 12a showed the highest propensity to label intracellular β2ARs in HEK293 T cell expressing exogenous β2ARs. This suggests that a combination of the polyethylene glycol (PEG) linker and the BODIPY‐X‐630/650 makes this ICI 118,551 derivative particularly susceptible to crossing the cell membrane to access the intracellular β2ARs. We have also used these ligands in combination with CRISPR/Cas9 genome‐edited HEK293 T cells to undertake for the first time real‐time ligand binding to native HEK293 T β2ARs at low native receptor expression levels. These studies provided quantitative data on ligand‐binding characteristics but also allowed real‐time visualization of the ligand‐binding interactions in genome‐edited cells using NanoBRET luminescence imaging.
Collapse
Affiliation(s)
- Joëlle Goulding
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Sarah J Mistry
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK.,School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Mark Soave
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Stephen J Briddon
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Carl W White
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK.,Harry Perkins Institute of Medical Research and Centre for Medical Research, University of Western Australia, QEII Medical Centre, Nedlands, Western Australia, Australia.,Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Barrie Kellam
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK.,School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| |
Collapse
|
9
|
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
|
10
|
Barkan K, Lagarias P, Stampelou M, Stamatis D, Hoare S, Safitri D, Klotz KN, Vrontaki E, Kolocouris A, Ladds G. Pharmacological characterisation of novel adenosine A 3 receptor antagonists. Sci Rep 2020; 10:20781. [PMID: 33247159 PMCID: PMC7695835 DOI: 10.1038/s41598-020-74521-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022] Open
Abstract
The adenosine A3 receptor (A3R) belongs to a family of four adenosine receptor (AR) subtypes which all play distinct roles throughout the body. A3R antagonists have been described as potential treatments for numerous diseases including asthma. Given the similarity between (adenosine receptors) orthosteric binding sites, obtaining highly selective antagonists is a challenging but critical task. Here we screen 39 potential A3R, antagonists using agonist-induced inhibition of cAMP. Positive hits were assessed for AR subtype selectivity through cAMP accumulation assays. The antagonist affinity was determined using Schild analysis (pA2 values) and fluorescent ligand binding. Structure-activity relationship investigations revealed that loss of the 3-(dichlorophenyl)-isoxazolyl moiety or the aromatic nitrogen heterocycle with nitrogen at α-position to the carbon of carboximidamide group significantly attenuated K18 antagonistic potency. Mutagenic studies supported by molecular dynamic simulations combined with Molecular Mechanics-Poisson Boltzmann Surface Area calculations identified the residues important for binding in the A3R orthosteric site. We demonstrate that K18, which contains a 3-(dichlorophenyl)-isoxazole group connected through carbonyloxycarboximidamide fragment with a 1,3-thiazole ring, is a specific A3R (< 1 µM) competitive antagonist. Finally, we introduce a model that enables estimates of the equilibrium binding affinity for rapidly disassociating compounds from real-time fluorescent ligand-binding studies. These results demonstrate the pharmacological characterisation of a selective competitive A3R antagonist and the description of its orthosteric binding mode. Our findings may provide new insights for drug discovery.
Collapse
Affiliation(s)
- Kerry Barkan
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Panagiotis Lagarias
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771, Athens, Greece
| | - Margarita Stampelou
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771, Athens, Greece
| | - Dimitrios Stamatis
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771, Athens, Greece
| | - Sam Hoare
- Pharmechanics LLC, 14 Sunnyside Drive South, Owego, NY, 13827, USA
| | - Dewi Safitri
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
- Pharmacology and Clinical Pharmacy Research Group, School of Pharmacy, Bandung Institute of Technology, Bandung, 40534, Indonesia
| | - Karl-Norbert Klotz
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078, Würzburg, Germany
| | - Eleni Vrontaki
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771, Athens, Greece
| | - Antonios Kolocouris
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771, Athens, Greece.
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK.
| |
Collapse
|
11
|
Comeo E, Kindon ND, Soave M, Stoddart LA, Kilpatrick LE, Scammells PJ, Hill SJ, Kellam B. Subtype-Selective Fluorescent Ligands as Pharmacological Research Tools for the Human Adenosine A 2A Receptor. J Med Chem 2020; 63:2656-2672. [PMID: 31887252 DOI: 10.1021/acs.jmedchem.9b01856] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Among class A G protein-coupled receptors (GPCR), the human adenosine A2A receptor (hA2AAR) remains an attractive drug target. However, translation of A2AAR ligands into the clinic has proved challenging and an improved understanding of A2AAR pharmacology could promote development of more efficacious therapies. Subtype-selective fluorescent probes would allow detailed real-time pharmacological investigations both in vitro and in vivo. In the present study, two families of fluorescent probes were designed around the known hA2AAR selective antagonist preladenant (SCH 420814). Both families of fluorescent antagonists retained affinity at the hA2AAR, selectivity over all other adenosine receptor subtypes and allowed clear visualization of specific receptor localization through confocal imaging. Furthermore, the Alexa Fluor 647-labeled conjugate allowed measurement of ligand binding affinities of unlabeled hA2AAR antagonists using a bioluminescence resonance energy transfer (NanoBRET) assay. The fluorescent ligands developed here can therefore be applied to a range of fluorescence-based techniques to further interrogate hA2AAR pharmacology and signaling.
Collapse
Affiliation(s)
- Eleonora Comeo
- Division of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, U.K.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands NG7 2UH, U.K.,Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052 Australia
| | - Nicholas D Kindon
- Division of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, U.K.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands NG7 2UH, U.K
| | - Mark Soave
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, U.K.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands NG7 2UH, U.K
| | - Leigh A Stoddart
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, U.K.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands NG7 2UH, U.K
| | - Laura E Kilpatrick
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, U.K.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands NG7 2UH, U.K
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052 Australia
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, U.K.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands NG7 2UH, U.K
| | - Barrie Kellam
- Division of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, U.K.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands NG7 2UH, U.K
| |
Collapse
|
12
|
Chemical Probes for the Adenosine Receptors. Pharmaceuticals (Basel) 2019; 12:ph12040168. [PMID: 31726680 PMCID: PMC6958474 DOI: 10.3390/ph12040168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 12/21/2022] Open
Abstract
Research on the adenosine receptors has been supported by the continuous discovery of new chemical probes characterized by more and more affinity and selectivity for the single adenosine receptor subtypes (A1, A2A, A2B and A3 adenosine receptors). Furthermore, the development of new techniques for the detection of G protein-coupled receptors (GPCR) requires new specific probes. In fact, if in the past radioligands were the most important GPCR probes for detection, compound screening and diagnostic purposes, nowadays, increasing importance is given to fluorescent and covalent ligands. In fact, advances in techniques such as fluorescence resonance energy transfer (FRET) and fluorescent polarization, as well as new applications in flow cytometry and different fluorescence-based microscopic techniques, are at the origin of the extensive research of new fluorescent ligands for these receptors. The resurgence of covalent ligands is due in part to a change in the common thinking in the medicinal chemistry community that a covalent drug is necessarily more toxic than a reversible one, and in part to the useful application of covalent ligands in GPCR structural biology. In this review, an updated collection of available chemical probes targeting adenosine receptors is reported.
Collapse
|
13
|
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
|
14
|
Dale NC, Johnstone EKM, White CW, Pfleger KDG. NanoBRET: The Bright Future of Proximity-Based Assays. Front Bioeng Biotechnol 2019; 7:56. [PMID: 30972335 PMCID: PMC6443706 DOI: 10.3389/fbioe.2019.00056] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/04/2019] [Indexed: 12/17/2022] Open
Abstract
Bioluminescence resonance energy transfer (BRET) is a biophysical technique used to monitor proximity within live cells. BRET exploits the naturally occurring phenomenon of dipole-dipole energy transfer from a donor enzyme (luciferase) to an acceptor fluorophore following enzyme-mediated oxidation of a substrate. This results in production of a quantifiable signal that denotes proximity between proteins and/or molecules tagged with complementary luciferase and fluorophore partners. BRET assays have been used to observe an array of biological functions including ligand binding, intracellular signaling, receptor-receptor proximity, and receptor trafficking, however, BRET assays can theoretically be used to monitor the proximity of any protein or molecule for which appropriate fusion constructs and/or fluorophore conjugates can be produced. Over the years, new luciferases and approaches have been developed that have increased the potential applications for BRET assays. In particular, the development of the small, bright and stable Nanoluciferase (NanoLuc; Nluc) and its use in NanoBRET has vastly broadened the potential applications of BRET assays. These advances have exciting potential to produce new experimental methods to monitor protein-protein interactions (PPIs), protein-ligand interactions, and/or molecular proximity. In addition to NanoBRET, Nluc has also been exploited to produce NanoBiT technology, which further broadens the scope of BRET to monitor biological function when NanoBiT is combined with an acceptor. BRET has proved to be a powerful tool for monitoring proximity and interaction, and these recent advances further strengthen its utility for a range of applications.
Collapse
Affiliation(s)
- Natasha C Dale
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia
| | - Elizabeth K M Johnstone
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia
| | - Carl W White
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia
| | - Kevin D G Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia.,Dimerix Limited, Nedlands, WA, Australia
| |
Collapse
|
15
|
Cooper AG, Oyagawa CRM, Manning JJ, Singh S, Hook S, Grimsey NL, Glass M, Tyndall JDA, Vernall AJ. Development of selective, fluorescent cannabinoid type 2 receptor ligands based on a 1,8-naphthyridin-2-(1 H)-one-3-carboxamide scaffold. MEDCHEMCOMM 2018; 9:2055-2067. [PMID: 30647881 PMCID: PMC6301273 DOI: 10.1039/c8md00448j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/18/2018] [Indexed: 12/16/2022]
Abstract
Cannabinoid type 2 (CB2) receptor has been implicated in several diseases and conditions, however no CB2 receptor selective drugs have made it to market. The aim of this study was to develop fluorescent ligands as CB2 receptor tools, to enable an increased understanding of CB2 receptor expression and signalling and thereby accelerate drug discovery. Fluorescent ligands have been successfully developed for other receptors, however none with adequate subtype selectivity or imaging properties have been reported for CB2 receptor. A series of 1,8-naphthyridin-2-(1H)-one-3-carboxamides with linkers and fluorophores appended in the N1 and C3-positions were developed. Molecular modelling indicated the C3 cis-cyclohexanol-linked compounds directed the linker out of the CB2 receptor between transmembrane helices 1 and 7. Herein we report fluorescent ligand 32 (hCB2 pK i = 6.33 ± 0.02) as one of the highest affinity, selective CB2 receptor fluorescent ligands reported. Despite 32 displaying poor specific labelling of CB2 receptor, the naphthyridine scaffold with this linker remains highly promising for future development of CB2 receptor tools.
Collapse
Affiliation(s)
- Anna G Cooper
- School of Pharmacy , University of Otago , 18 Frederick Street , Dunedin 9054 , New Zealand . ; Tel: +64 3 479 4518
| | - Caitlin R M Oyagawa
- Department of Pharmacology and Clinical Pharmacology, and Centre for Brain Research , School of Medical Sciences , University of Auckland , Auckland , New Zealand
| | - Jamie J Manning
- Department of Pharmacology and Clinical Pharmacology, and Centre for Brain Research , School of Medical Sciences , University of Auckland , Auckland , New Zealand
| | - Sameek Singh
- School of Pharmacy , University of Otago , 18 Frederick Street , Dunedin 9054 , New Zealand . ; Tel: +64 3 479 4518
| | - Sarah Hook
- School of Pharmacy , University of Otago , 18 Frederick Street , Dunedin 9054 , New Zealand . ; Tel: +64 3 479 4518
| | - Natasha L Grimsey
- Department of Pharmacology and Clinical Pharmacology, and Centre for Brain Research , School of Medical Sciences , University of Auckland , Auckland , New Zealand
| | - Michelle Glass
- Department of Pharmacology and Clinical Pharmacology, and Centre for Brain Research , School of Medical Sciences , University of Auckland , Auckland , New Zealand
| | - Joel D A Tyndall
- School of Pharmacy , University of Otago , 18 Frederick Street , Dunedin 9054 , New Zealand . ; Tel: +64 3 479 4518
| | - Andrea J Vernall
- School of Pharmacy , University of Otago , 18 Frederick Street , Dunedin 9054 , New Zealand . ; Tel: +64 3 479 4518
| |
Collapse
|
16
|
Iliopoulos-Tsoutsouvas C, Kulkarni RN, Makriyannis A, Nikas SP. Fluorescent probes for G-protein-coupled receptor drug discovery. Expert Opin Drug Discov 2018; 13:933-947. [DOI: 10.1080/17460441.2018.1518975] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | - Rohit N. Kulkarni
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center. Department of Medicine, Brigham and Women’s Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery and Departments of Chemistry & Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Spyros P. Nikas
- Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Köse M, Gollos S, Karcz T, Fiene A, Heisig F, Behrenswerth A, Kieć-Kononowicz K, Namasivayam V, Müller CE. Fluorescent-Labeled Selective Adenosine A 2B Receptor Antagonist Enables Competition Binding Assay by Flow Cytometry. J Med Chem 2018; 61:4301-4316. [PMID: 29681156 DOI: 10.1021/acs.jmedchem.7b01627] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fluorescent ligands represent powerful tools for biological studies and are considered attractive alternatives to radioligands. In this study, we developed fluorescent antagonists for A2B adenosine receptors (A2BARs), which are targeted by antiasthmatic xanthines and were proposed as novel targets in immuno-oncology. Our approach was to merge a small borondipyrromethene (BODIPY) derivative with the pharmacophore of 8-substituted xanthine derivatives. On the basis of the design, synthesis, and evaluation of model compounds, several fluorescent ligands were synthesized. Compound 29 (PSB-12105), which displayed high affinity for human, rat, and mouse A2BARs ( Ki = 0.2-2 nM) and high selectivity for this AR subtype, was selected for further studies. A homology model of the human A2BAR was generated, and docking studies were performed. Moreover, 29 allowed us to establish a homogeneous receptor-ligand binding assay using flow cytometry. These compounds constitute the first potent, selective fluorescent A2BAR ligands and are anticipated to be useful for a variety of applications.
Collapse
Affiliation(s)
- Meryem Köse
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I , University of Bonn , An der Immenburg 4 , D-53121 Bonn , Germany
| | - Sabrina Gollos
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I , University of Bonn , An der Immenburg 4 , D-53121 Bonn , Germany
| | - Tadeusz Karcz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy , Jagiellonian University Medical College , Medyczna 9 , 30-688 Kraków , Poland
| | - Amelie Fiene
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I , University of Bonn , An der Immenburg 4 , D-53121 Bonn , Germany
| | - Fabian Heisig
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I , University of Bonn , An der Immenburg 4 , D-53121 Bonn , Germany
| | - Andrea Behrenswerth
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I , University of Bonn , An der Immenburg 4 , D-53121 Bonn , Germany
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy , Jagiellonian University Medical College , Medyczna 9 , 30-688 Kraków , Poland
| | - Vigneshwaran Namasivayam
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I , University of Bonn , An der Immenburg 4 , D-53121 Bonn , Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I , University of Bonn , An der Immenburg 4 , D-53121 Bonn , Germany
| |
Collapse
|
18
|
Lee S, Cil O, Diez-Cecilia E, Anderson MO, Verkman AS. Nanomolar-Potency 1,2,4-Triazoloquinoxaline Inhibitors of the Kidney Urea Transporter UT-A1. J Med Chem 2018; 61:3209-3217. [PMID: 29589443 PMCID: PMC5976253 DOI: 10.1021/acs.jmedchem.8b00343] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Urea transporter A (UT-A) isoforms encoded by the Slc14a2 gene are expressed in kidney tubule epithelial cells, where they facilitate urinary concentration. UT-A1 inhibition is predicted to produce a unique salt-sparing diuretic action in edema and hyponatremia. Here we report the discovery of 1,2,4-triazoloquinoxalines and the analysis of 37 synthesized analogues. The most potent compound, 8ay, containing 1,2,4-triazolo[4,3- a]quinoxaline-substituted benzenesulfonamide linked by an aryl ether, rapidly and reversibly inhibited UT-A1 urea transport by a noncompetitive mechanism with IC50 ≈ 150 nM; the IC50 was ∼2 μM for the related urea transporter UT-B encoded by the Slc14a1 gene. Molecular modeling suggested a putative binding site on the UT-A1 cytoplasmic domain. In vitro metabolism showing quinoxaline ring oxidation prompted the synthesis of metabolically stable 7,8-difluoroquinoxaline analogue 8bl, which when administered to rats produced marked diuresis and reduced urinary osmolality. 8bl has substantially improved UT-A1 inhibition potency and metabolic stability compared with prior compounds.
Collapse
Affiliation(s)
- Sujin Lee
- Departments of Medicine and Physiology, University of California, San Francisco, California 94143-0521, United States
| | - Onur Cil
- Departments of Medicine and Physiology, University of California, San Francisco, California 94143-0521, United States
- Department of Pediatrics, Division of Nephrology, University of California, San Francisco, California 94143-0521, United States
| | - Elena Diez-Cecilia
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, California 94132-1722, United States
| | - Marc O. Anderson
- Departments of Medicine and Physiology, University of California, San Francisco, California 94143-0521, United States
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, California 94132-1722, United States
| | - Alan S. Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, California 94143-0521, United States
| |
Collapse
|
19
|
Peiró Cadahía J, Bondebjerg J, Hansen CA, Previtali V, Hansen AE, Andresen TL, Clausen MH. Synthesis and Evaluation of Hydrogen Peroxide Sensitive Prodrugs of Methotrexate and Aminopterin for the Treatment of Rheumatoid Arthritis. J Med Chem 2018; 61:3503-3515. [PMID: 29605999 DOI: 10.1021/acs.jmedchem.7b01775] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A series of novel hydrogen peroxide sensitive prodrugs of methotrexate (MTX) and aminopterin (AMT) were synthesized and evaluated for therapeutic efficacy in mice with collagen induced arthritis (CIA) as a model of chronic rheumatoid arthritis (RA). The prodrug strategy selected is based on ROS-labile 4-methylphenylboronic acid promoieties linked to the drugs via a carbamate linkage or a direct C-N bond. Activation under pathophysiological concentrations of H2O2 proved to be effective, and prodrug candidates were selected in agreement with relevant in vitro physicochemical and pharmacokinetic assays. Selected candidates showed moderate to good solubility, high chemical and enzymatic stability, and therapeutic efficacy comparable to the parent drugs in the CIA model. Importantly, the prodrugs displayed the expected safer toxicity profile and increased therapeutic window compared to MTX and AMT while maintaining a comparable therapeutic efficacy, which is highly encouraging for future use in RA patients.
Collapse
Affiliation(s)
- Jorge Peiró Cadahía
- Center for Nanomedicine & Theranostics, Department of Chemistry , Technical University of Denmark , Kemitorvet 207 , DK-2800 Kongens Lyngby , Denmark
| | - Jon Bondebjerg
- MC2 Therapeutics , Agern Alle 24-26 , 2970 Hørsholm , Denmark
| | | | - Viola Previtali
- Center for Nanomedicine & Theranostics, Department of Chemistry , Technical University of Denmark , Kemitorvet 207 , DK-2800 Kongens Lyngby , Denmark
| | - Anders E Hansen
- Center for Nanomedicine & Theranostics, Department of Micro- and Nanotechnology , Technical University of Denmark , Ørsteds Plads, Building 345 , DK-2800 Kongens Lyngby , Denmark
| | - Thomas L Andresen
- Center for Nanomedicine & Theranostics, Department of Micro- and Nanotechnology , Technical University of Denmark , Ørsteds Plads, Building 345 , DK-2800 Kongens Lyngby , Denmark
| | - Mads H Clausen
- Center for Nanomedicine & Theranostics, Department of Chemistry , Technical University of Denmark , Kemitorvet 207 , DK-2800 Kongens Lyngby , Denmark
| |
Collapse
|
20
|
Stoddart LA, Kilpatrick LE, Hill SJ. NanoBRET Approaches to Study Ligand Binding to GPCRs and RTKs. Trends Pharmacol Sci 2018; 39:136-147. [PMID: 29132917 DOI: 10.1016/j.tips.2017.10.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/12/2017] [Accepted: 10/17/2017] [Indexed: 12/30/2022]
Abstract
Recent advances in the development of fluorescent ligands for G-protein-coupled receptors (GPCRs) and receptor tyrosine kinase receptors (RTKs) have facilitated the study of these receptors in living cells. A limitation of these ligands is potential uptake into cells and increased nonspecific binding. However, this can largely be overcome by using proximity approaches, such as bioluminescence resonance energy transfer (BRET), which localise the signal (within 10nm) to the specific receptor target. The recent engineering of NanoLuc has resulted in a luciferase variant that is smaller and significantly brighter (up to tenfold) than existing variants. Here, we review the use of BRET from N-terminal NanoLuc-tagged GPCRs or a RTK to a receptor-bound fluorescent ligand to provide quantitative pharmacology of ligand-receptor interactions in living cells in real time.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Division of Physiology, Pharmacology 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, The Midlands, UK; These authors contributed equally to this work
| | - Laura E Kilpatrick
- Division of Physiology, Pharmacology 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, The Midlands, UK; These authors contributed equally to this work
| | - Stephen J Hill
- Division of Physiology, Pharmacology 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, The Midlands, UK.
| |
Collapse
|
21
|
Cooper AG, MacDonald C, Glass M, Hook S, Tyndall JD, Vernall AJ. Alkyl indole-based cannabinoid type 2 receptor tools: Exploration of linker and fluorophore attachment. Eur J Med Chem 2018; 145:770-789. [DOI: 10.1016/j.ejmech.2017.11.076] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 11/23/2017] [Accepted: 11/26/2017] [Indexed: 01/03/2023]
|
22
|
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
|
23
|
Lam R, Gondin AB, Canals M, Kellam B, Briddon SJ, Graham B, Scammells PJ. Fluorescently Labeled Morphine Derivatives for Bioimaging Studies. J Med Chem 2018; 61:1316-1329. [DOI: 10.1021/acs.jmedchem.7b01811] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Raymond Lam
- School
of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Arisbel B. Gondin
- Cell Signalling
Research Group, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, U.K
| | | | - Barrie Kellam
- School
of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Stephen J. Briddon
- Cell Signalling
Research Group, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, U.K
| | | | | |
Collapse
|
24
|
Arruda MA, Stoddart LA, Gherbi K, Briddon SJ, Kellam B, Hill SJ. A Non-imaging High Throughput Approach to Chemical Library Screening at the Unmodified Adenosine-A 3 Receptor in Living Cells. Front Pharmacol 2017; 8:908. [PMID: 29321740 PMCID: PMC5733478 DOI: 10.3389/fphar.2017.00908] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/28/2017] [Indexed: 12/29/2022] Open
Abstract
Recent advances in fluorescent ligand technology have enabled the study of G protein-coupled receptors in their native environment without the need for genetic modification such as addition of N-terminal fluorescent or bioluminescent tags. Here, we have used a non-imaging plate reader (PHERAstar FS) to monitor the binding of fluorescent ligands to the human adenosine-A3 receptor (A3AR; CA200645 and AV039), stably expressed in CHO-K1 cells. To verify that this method was suitable for the study of other GPCRs, assays at the human adenosine-A1 receptor, and β1 and β2 adrenoceptors (β1AR and β2AR; BODIPY-TMR-CGP-12177) were also carried out. Affinity values determined for the binding of the fluorescent ligands CA200645 and AV039 to A3AR for a range of classical adenosine receptor antagonists were consistent with A3AR pharmacology and correlated well (R2 = 0.94) with equivalent data obtained using a confocal imaging plate reader (ImageXpress Ultra). The binding of BODIPY-TMR-CGP-12177 to the β1AR was potently inhibited by low concentrations of the β1-selective antagonist CGP 20712A (pKi 9.68) but not by the β2-selective antagonist ICI 118551(pKi 7.40). Furthermore, in experiments conducted in CHO K1 cells expressing the β2AR this affinity order was reversed with ICI 118551 showing the highest affinity (pKi 8.73) and CGP20712A (pKi 5.68) the lowest affinity. To determine whether the faster data acquisition of the non-imaging plate reader (~3 min per 96-well plate) was suitable for high throughput screening (HTS), we screened the LOPAC library for inhibitors of the binding of CA200645 to the A3AR. From the initial 1,263 compounds evaluated, 67 hits (defined as those that inhibited the total binding of 25 nM CA200645 by ≥40%) were identified. All compounds within the library that had medium to high affinity for the A3AR (pKi ≥6) were successfully identified. We found three novel compounds in the library that displayed unexpected sub-micromolar affinity for the A3AR. These were K114 (pKi 6.43), retinoic acid p-hydroxyanilide (pKi 6.13) and SU 6556 (pKi 6.17). Molecular docking of these latter three LOPAC library members provided a plausible set of binding poses within the vicinity of the established orthosteric A3AR binding pocket. A plate reader based library screening using an untagged receptor is therefore possible using fluorescent ligand opening the possibility of its use in compound screening at natively expressed receptors.
Collapse
Affiliation(s)
- Maria Augusta Arruda
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom
- Vice-Diretoria de Ensino, Pesquisa e Inovacao, Farmanguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Leigh A. Stoddart
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom
| | - Karolina Gherbi
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom
| | - Stephen J. Briddon
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom
| | - Barrie Kellam
- Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Stephen J. Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom
| |
Collapse
|
25
|
High-Affinity Functional Fluorescent Ligands for Human β-Adrenoceptors. Sci Rep 2017; 7:12319. [PMID: 28951558 PMCID: PMC5614969 DOI: 10.1038/s41598-017-12468-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/08/2017] [Indexed: 01/14/2023] Open
Abstract
Visualization of the G-protein coupled receptor (GPCR) is of great importance for studying its function in a native cell. We have synthesized a series of red-emitting fluorescent probes targeting β-adrenergic receptor (βAR) that are compatible with confocal and Stimulated Emission Depletion (STED) microscopy as well as with Time-Resolved Fluorescence Resonance Energy Transfer (TR-FRET) binding assay in living cells. The probe based on the agonist BI-167107 and fluorescent dye KK114 demonstrates nanomolar binding affinity and up to nine-fold β2AR selectivity over β1AR. Carazolol-derived probes are fluorogenic and allow no-wash imaging experiments. STED microscopy of β2ARs stained at the native expression level on pancreatic CAPAN cells provides two-fold improvement in lateral optical resolution over confocal mode and reveals the formation of receptor microdomains. These probes retain their functional (agonist or antagonist) properties, allowing simultaneous modulation of cyclic adenosine monophosphate (cAMP) levels and receptor internalization as well as imaging receptor localization.
Collapse
|
26
|
Schwehm C, Kellam B, Garces AE, Hill SJ, Kindon ND, Bradshaw TD, Li J, Macdonald SJF, Rowedder JE, Stoddart LA, Stocks MJ. Design and Elaboration of a Tractable Tricyclic Scaffold To Synthesize Druglike Inhibitors of Dipeptidyl Peptidase-4 (DPP-4), Antagonists of the C-C Chemokine Receptor Type 5 (CCR5), and Highly Potent and Selective Phosphoinositol-3 Kinase δ (PI3Kδ) Inhibitors. J Med Chem 2017; 60:1534-1554. [PMID: 28128944 DOI: 10.1021/acs.jmedchem.6b01801] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A novel molecular scaffold has been synthesized, and its incorporation into new analogues of biologically active molecules across multiple target classes will be discussed. In these studies, we have shown use of the tricyclic scaffold to synthesize potent inhibitors of the serine peptidase DPP-4, antagonists of the CCR5 receptor, and highly potent and selective PI3K δ isoform inhibitors. We also describe the predicted physicochemical properties of the resulting inhibitors and conclude that the tractable molecular scaffold could have potential application in future drug discovery programs.
Collapse
Affiliation(s)
- Carolin Schwehm
- School of Pharmacy, Centre for Biomolecular Sciences, University Park Nottingham , Nottingham, NG7 2RD, U.K
| | - Barrie Kellam
- School of Pharmacy, Centre for Biomolecular Sciences, University Park Nottingham , Nottingham, NG7 2RD, U.K
| | - Aimie E Garces
- School of Pharmacy, Centre for Biomolecular Sciences, University Park Nottingham , Nottingham, NG7 2RD, U.K
| | - Stephen J Hill
- Institute of Cell Signalling, Medical School, University of Nottingham , Nottingham, NG7 2UH, U.K
| | - Nicholas D Kindon
- School of Pharmacy, Centre for Biomolecular Sciences, University Park Nottingham , Nottingham, NG7 2RD, U.K
| | - Tracey D Bradshaw
- School of Pharmacy, Centre for Biomolecular Sciences, University Park Nottingham , Nottingham, NG7 2RD, U.K
| | - Jin Li
- Hitgen Ltd. , F7-10, Building B3, Tianfu Life Science Park, 88 South Kayuan Road, Chengdu, Sichuan, China 610041
| | - Simon J F Macdonald
- GlaxoSmithKline , Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, U.K
| | - James E Rowedder
- GlaxoSmithKline , Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, U.K
| | - Leigh A Stoddart
- Institute of Cell Signalling, Medical School, University of Nottingham , Nottingham, NG7 2UH, U.K
| | - Michael J Stocks
- School of Pharmacy, Centre for Biomolecular Sciences, University Park Nottingham , Nottingham, NG7 2RD, U.K
| |
Collapse
|
27
|
Ranganathan A, Stoddart LA, Hill SJ, Carlsson J. Fragment-Based Discovery of Subtype-Selective Adenosine Receptor Ligands from Homology Models. J Med Chem 2015; 58:9578-90. [PMID: 26592528 DOI: 10.1021/acs.jmedchem.5b01120] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fragment-based lead discovery (FBLD) holds great promise for drug discovery, but applications to G protein-coupled receptors (GPCRs) have been limited by a lack of sensitive screening techniques and scarce structural information. If virtual screening against homology models of GPCRs could be used to identify fragment ligands, FBLD could be extended to numerous important drug targets and contribute to efficient lead generation. Access to models of multiple receptors may further enable the discovery of fragments that bind specifically to the desired target. To investigate these questions, we used molecular docking to screen >500 000 fragments against homology models of the A3 and A1 adenosine receptors (ARs) with the goal to discover A3AR-selective ligands. Twenty-one fragments with predicted A3AR-specific binding were evaluated in live-cell fluorescence-based assays; of eight verified ligands, six displayed A3/A1 selectivity, and three of these had high affinities ranging from 0.1 to 1.3 μM. Subsequently, structure-guided fragment-to-lead optimization led to the identification of a >100-fold-selective antagonist with nanomolar affinity from commercial libraries. These results highlight that molecular docking screening can guide fragment-based discovery of selective ligands even if the structures of both the target and antitarget receptors are unknown. The same approach can be readily extended to a large number of pharmaceutically important targets.
Collapse
Affiliation(s)
- Anirudh Ranganathan
- Science for Life Laboratory, Department of Biochemistry and Biophysics, and Center for Biomembrane Research, Stockholm University , SE-106 91 Stockholm, Sweden
| | - Leigh A Stoddart
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham , Nottingham NG7 2UH, U.K
| | - Stephen J Hill
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham , Nottingham NG7 2UH, U.K
| | - Jens Carlsson
- Science for Life Laboratory, Department of Medicinal Chemistry, BMC, Uppsala University , P.O. Box 574, SE-751 23 Uppsala, Sweden
| |
Collapse
|
28
|
Gherbi K, Briddon SJ, Hill SJ. Detection of the secondary, low-affinity β1 -adrenoceptor site in living cells using the fluorescent CGP 12177 derivative BODIPY-TMR-CGP. Br J Pharmacol 2015; 171:5431-45. [PMID: 25052258 PMCID: PMC4261997 DOI: 10.1111/bph.12858] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 07/11/2014] [Accepted: 07/18/2014] [Indexed: 02/06/2023] Open
Abstract
Background and Purpose CGP 12177 not only inhibits agonist effects mediated through the catecholamine site of the β1-adrenoceptor with high affinity, but also exhibits agonist effects of its own at higher concentrations through a secondary, low-affinity β1-adrenoceptor site or conformation. β-blocker affinities for this ‘CGP 12177’ site of the human β1-adrenoceptor have thus far only been characterized in functional studies. Here, we used the fluorescent CGP 12177 analogue BODIPY-TMR-CGP to directly investigate receptor–ligand interactions at the secondary binding site of the β1-adrenoceptor. Experimental Approach The human β1-adrenoceptor was stably expressed in CHO cells containing a cAMP response element (CRE)-secreted placental alkaline phosphatase (SPAP) reporter gene construct. Functional responses of BODIPY-TMR-CGP were determined in the CRE-SPAP reporter gene assay, and manual and automated confocal microscopy platforms used to investigate the binding properties of BODIPY-TMR-CGP. Key Results BODIPY-TMR-CGP displayed a pharmacological profile similar to that of CGP 12177, retaining agonist activity at the secondary β1-adrenoceptor site. In confocal microscopy studies, specific BODIPY-TMR-CGP binding allowed clear visualization of β1-adrenoceptors in live cells. Using a wider concentration range of labelled ligand in a high-content fluorescence-based binding assay than is possible in radioligand binding assays, two-site inhibition binding curves of β-adrenoceptor antagonists were revealed in CHO cells expressing the human β1-adrenoceptor, but not the β2-adrenoceptor. Conclusions and Implications The fluorescent CGP 12177 analogue allowed the detection of the β1-adrenoceptor secondary site in both functional and binding studies. This suggests that BODIPY-TMR-CGP presents an important and novel fluorescent tool to investigate the nature of the secondary β1-adrenoceptor site.
Collapse
Affiliation(s)
- K Gherbi
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | | |
Collapse
|
29
|
Stoddart LA, Johnstone EKM, Wheal AJ, Goulding J, Robers MB, Machleidt T, Wood KV, Hill SJ, Pfleger KDG. Application of BRET to monitor ligand binding to GPCRs. Nat Methods 2015; 12:661-663. [PMID: 26030448 PMCID: PMC4488387 DOI: 10.1038/nmeth.3398] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/04/2015] [Indexed: 01/17/2023]
Abstract
Bioluminescence resonance energy transfer (BRET) is a well-established method for investigating protein-protein interactions. Here we present a novel BRET approach to monitor ligand binding to G protein-coupled receptors (GPCRs) on the surface of living cells made possible by the use of fluorescent ligands in combination with a novel bioluminescent protein (NanoLuc) that can be readily expressed on the N-terminus of GPCRs.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham Medical School, Nottingham, United Kingdom
| | - Elizabeth K M Johnstone
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Amanda J Wheal
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham Medical School, Nottingham, United Kingdom
| | - Joëlle Goulding
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham Medical School, Nottingham, United Kingdom
| | | | | | - Keith V Wood
- Promega Corporation, Madison, Wisconsin, United States
| | - Stephen J Hill
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham Medical School, Nottingham, United Kingdom.,Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Kevin D G Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
30
|
Stoddart LA, Kilpatrick LE, Briddon SJ, Hill SJ. Probing the pharmacology of G protein-coupled receptors with fluorescent ligands. Neuropharmacology 2015; 98:48-57. [PMID: 25979488 DOI: 10.1016/j.neuropharm.2015.04.033] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/23/2015] [Accepted: 04/29/2015] [Indexed: 01/01/2023]
Abstract
G protein-coupled receptors control a wide range of physiological processes and are the target for many clinically used drugs. Understanding the way in which receptors bind agonists and antagonists, their organisation in the membrane and their regulation after agonist binding are important properties which are key to developing new drugs. One way to achieve this knowledge is through the use of fluorescent ligands, which have been used to study the expression and function of receptors in endogenously expressing systems. Fluorescent ligands with appropriate imaging properties can be used in conjunction with confocal microscopy to investigate the regulation of receptors after activation. Alternatively, through the use of single molecule microscopy, they can probe the spatial organisation of receptors within the membrane. This review focuses on the techniques in which fluorescent ligands have been used and the novel aspects of G protein-coupled receptor pharmacology which have been uncovered. This article is part of the Special Issue entitled 'Fluorescent Tools in Neuropharmacology'.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Laura E Kilpatrick
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Stephen J Briddon
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Stephen J Hill
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
31
|
Stoddart LA, Vernall AJ, Briddon SJ, Kellam B, Hill SJ. Direct visualisation of internalization of the adenosine A3 receptor and localization with arrestin3 using a fluorescent agonist. Neuropharmacology 2015; 98:68-77. [PMID: 25937210 DOI: 10.1016/j.neuropharm.2015.04.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/19/2015] [Accepted: 04/14/2015] [Indexed: 11/25/2022]
Abstract
Fluorescence based probes provide a novel way to study the dynamic internalization process of G protein-coupled receptors (GPCRs). Recent advances in the rational design of fluorescent ligands for GPCRs have been used here to generate new fluorescent agonists containing tripeptide linkers for the adenosine A3 receptor. The fluorescent agonist BY630-X-(D)-A-(D)-A-G-ABEA was found to be a highly potent agonist at the adenosine A3 receptor in both reporter gene (pEC50 = 8.48 ± 0.09) and internalization assays (pEC50 = 7.47 ± 0.11). Confocal imaging studies showed that BY630-X-(D)-A-(D)-A-G-ABEA was internalized with A3 linked to yellow fluorescent protein, which was blocked by the competitive antagonist MRS1220. Internalization of untagged adenosine A3 could also be visualized with BY630-X-(D)-A-(D)-A-G-ABEA treatment. Further, BY630-X-(D)-A-(D)-A-G-ABEA stimulated the formation of receptor-arrestin3 complexes and was found to localize with these intracellular complexes. This highly potent agonist with excellent imaging properties should be a valuable tool to study receptor internalization. This article is part of the Special Issue entitled 'Fluorescent Tools in Neuropharmacology'.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Cell Signalling Research Group, School of Life Sciences, Queen's Medical Centre, University of Nottingham, NG7 2UH, UK
| | - Andrea J Vernall
- School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Stephen J Briddon
- Cell Signalling Research Group, School of Life Sciences, Queen's Medical Centre, University of Nottingham, NG7 2UH, UK
| | - Barrie Kellam
- School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Stephen J Hill
- Cell Signalling Research Group, School of Life Sciences, Queen's Medical Centre, University of Nottingham, NG7 2UH, UK.
| |
Collapse
|
32
|
Sridharan R, Zuber J, Connelly SM, Mathew E, Dumont ME. Fluorescent approaches for understanding interactions of ligands with G protein coupled receptors. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1838:15-33. [PMID: 24055822 PMCID: PMC3926105 DOI: 10.1016/j.bbamem.2013.09.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 09/03/2013] [Accepted: 09/08/2013] [Indexed: 11/18/2022]
Abstract
G protein coupled receptors are responsible for a wide variety of signaling responses in diverse cell types. Despite major advances in the determination of structures of this class of receptors, the underlying mechanisms by which binding of different types of ligands specifically elicits particular signaling responses remain unclear. The use of fluorescence spectroscopy can provide important information about the process of ligand binding and ligand dependent conformational changes in receptors, especially kinetic aspects of these processes that can be difficult to extract from X-ray structures. We present an overview of the extensive array of fluorescent ligands that have been used in studies of G protein coupled receptors and describe spectroscopic approaches for assaying binding and probing the environment of receptor-bound ligands with particular attention to examples involving yeast pheromone receptors. In addition, we discuss the use of fluorescence spectroscopy for detecting and characterizing conformational changes in receptors induced by the binding of ligands. Such studies have provided strong evidence for diversity of receptor conformations elicited by different ligands, consistent with the idea that GPCRs are not simple on and off switches. This diversity of states constitutes an underlying mechanistic basis for biased agonism, the observation that different stimuli can produce different responses from a single receptor. It is likely that continued technical advances will allow fluorescence spectroscopy to play an important role in continued probing of structural transitions in G protein coupled receptors. This article is part of a Special Issue entitled: Structural and biophysical characterisation of membrane protein-ligand binding.
Collapse
Affiliation(s)
- Rajashri Sridharan
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Jeffrey Zuber
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Sara M. Connelly
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Elizabeth Mathew
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Mark E. Dumont
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pediatrics, P.O. Box 777, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
33
|
Jaeger WC, Armstrong SP, Hill SJ, Pfleger KDG. Biophysical Detection of Diversity and Bias in GPCR Function. Front Endocrinol (Lausanne) 2014; 5:26. [PMID: 24634666 PMCID: PMC3943086 DOI: 10.3389/fendo.2014.00026] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 02/19/2014] [Indexed: 12/27/2022] Open
Abstract
Guanine nucleotide binding protein (G protein)-coupled receptors (GPCRs) function in complexes with a range of molecules and proteins including ligands, G proteins, arrestins, ubiquitin, and other receptors. Elements of these complexes may interact constitutively or dynamically, dependent upon factors such as ligand binding, phosphorylation, and dephosphorylation. They may also be allosterically modulated by other proteins in a manner that changes temporally and spatially within the cell. Elucidating how these complexes function has been greatly enhanced by biophysical technologies that are able to monitor proximity and/or binding, often in real time and in live cells. These include resonance energy transfer approaches such as bioluminescence resonance energy transfer (BRET) and fluorescence resonance energy transfer (FRET). Furthermore, the use of fluorescent ligands has enabled novel insights into allosteric interactions between GPCRs. Consequently, biophysical approaches are helping to unlock the amazing diversity and bias in G protein-coupled receptor signaling.
Collapse
Affiliation(s)
- Werner C. Jaeger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Stephen P. Armstrong
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Stephen J. Hill
- Cell Signalling Research Group, School of Life Sciences, Queen’s Medical Centre, University of Nottingham Medical School, Nottingham, UK
| | - Kevin D. G. Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- Dimerix Bioscience Pty Ltd, Perth, WA, Australia
- *Correspondence: Kevin D. G. Pfleger, Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, QEII Medical Centre, QQ Block, 6 Verdun Street, Nedlands, Perth, WA 6009, Australia e-mail:
| |
Collapse
|
34
|
Heisig F, Gollos S, Freudenthal SJ, El-Tayeb A, Iqbal J, Müller CE. Synthesis of BODIPY derivatives substituted with various bioconjugatable linker groups: a construction kit for fluorescent labeling of receptor ligands. J Fluoresc 2013; 24:213-30. [PMID: 24052460 DOI: 10.1007/s10895-013-1289-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 08/09/2013] [Indexed: 12/15/2022]
Abstract
The goal of the present study was to design small, functionalized green-emitting BODIPY dyes, which can readily be coupled to target molecules such as receptor ligands, or even be integrated into their pharmacophores. A simple two-step one-pot procedure starting from 2,4-dimethylpyrrole and ω-bromoalkylcarboxylic acid chlorides was used to obtain new ω-bromoalkyl-substituted BODIPY fluorophores (1a-1f) connected via alkyl spacers of different length to the 8-position of the fluorescent dye. The addition of radical inhibitors reduced the amount of side products. The ω-bromoalkyl-substituted BODIPYs were further converted to introduce various functional groups: iodo-substituted dyes were obtained by Finkelstein reaction in excellent yields; microwave-assisted reaction with methanolic ammonia led to fast and clean conversion to the amino-substituted dyes; a hydroxyl-substituted derivative was prepared by reaction with sodium ethylate, and thiol-substituted BODIPYs were obtained by reaction of 1a-1f with potassium thioacetate followed by alkaline cleavage of the thioesters. Water-soluble derivatives were prepared by introducing sulfonate groups into the 2- and 6-position of the BODIPY core. The synthesized BODIPY derivatives showed high fluorescent yields and appeared to be stable under basic, reducing and oxidative conditions. As a proof of concept, 2-thioadenosine was alkylated with bromoethyl-BODIPY 1b. The resulting fluorescent 2-substituted adenosine derivative 15 displayed selectivity for the A3 adenosine receptor (ARs) over the other AR subtypes, showed agonistic activity, and may thus become a useful tool for studying A3ARs, or a lead structure for further optimization. The new functionalized dyes may be widely used for fluorescent labeling allowing the investigation of biological targets and processes.
Collapse
Affiliation(s)
- Fabian Heisig
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University Bonn, 53121, Bonn, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Jörg M, Shonberg J, Mak FS, Miller ND, Yuriev E, Scammells PJ, Capuano B. Novel adenosine A2A receptor ligands: A synthetic, functional and computational investigation of selected literature adenosine A2A receptor antagonists for extending into extracellular space. Bioorg Med Chem Lett 2013; 23:3427-33. [DOI: 10.1016/j.bmcl.2013.03.070] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 03/15/2013] [Accepted: 03/20/2013] [Indexed: 11/16/2022]
|
36
|
Kozma E, Gizewski ET, Tosh DK, Squarcialupi L, Auchampach JA, Jacobson KA. Characterization by flow cytometry of fluorescent, selective agonist probes of the A(3) adenosine receptor. Biochem Pharmacol 2013; 85:1171-81. [PMID: 23376019 DOI: 10.1016/j.bcp.2013.01.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Revised: 01/16/2013] [Accepted: 01/24/2013] [Indexed: 01/02/2023]
Abstract
Various fluorescent nucleoside agonists of the A3 adenosine receptor (AR) were compared as high affinity probes using radioligands and flow cytometry (FCM). They contained a fluorophore linked through the C2 or N(6) position and rigid A3AR-enhancing (N)-methanocarba modification. A hydrophobic C2-(1-pyrenyl) derivative MRS5704 bound nonselectively. C2-Tethered cyanine5-dye labeled MRS5218 bound selectively to hA3AR expressed in whole CHO cells and membranes. By FCM, binding was A3AR-mediated (blocked by A3AR antagonist, at least half through internalization), with t1/2 for association 38min in mA3AR-HEK293 cells; 26.4min in sucrose-treated hA3AR-CHO cells (Kd 31nM). Membrane binding indicated moderate mA3AR affinity, but not selectivity. Specific accumulation of fluorescence (50nM MRS5218) occurred in cells expressing mA3AR, but not other mouse ARs. Evidence was provided suggesting that MRS5218 detects endogenous expression of the A3AR in the human promyelocytic leukemic HL-60 cell line. Therefore, MRS5218 promises to be a useful tool for characterizing the A3AR.
Collapse
Affiliation(s)
- Eszter Kozma
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0810, USA
| | | | | | | | | | | |
Collapse
|
37
|
Kozma E, Jayasekara PS, Squarcialupi L, Paoletta S, Moro S, Federico S, Spalluto G, Jacobson KA. Fluorescent ligands for adenosine receptors. Bioorg Med Chem Lett 2013; 23:26-36. [PMID: 23200243 PMCID: PMC3557833 DOI: 10.1016/j.bmcl.2012.10.112] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 10/05/2012] [Accepted: 10/28/2012] [Indexed: 10/27/2022]
Abstract
Interest is increasing in developing fluorescent ligands for characterization of adenosine receptors (ARs), which hold a promise of usefulness in the drug discovery process. The size of a strategically labeled AR ligand can be greatly increased after the attachment of a fluorophore. The choice of dye moiety (e.g. Alexa Fluor 488), attachment point and linker length can alter the selectivity and potency of the parent molecule. Fluorescent derivatives of adenosine agonists and antagonists (e.g. XAC and other heterocyclic antagonist scaffolds) have been synthesized and characterized pharmacologically. Some are useful AR probes for flow cytometry, fluorescence correlation spectroscopy, fluorescence microscopy, fluorescence polarization, fluorescence resonance energy transfer, and scanning confocal microscopy. Thus, the approach of fluorescent labeled GPCR ligands, including those for ARs, is a growing dynamic research field.
Collapse
Affiliation(s)
- Eszter Kozma
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810 USA
| | - P Suresh Jayasekara
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810 USA
| | - Lucia Squarcialupi
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810 USA
| | - Silvia Paoletta
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810 USA
| | - Stefano Moro
- Molecular Modeling Section (MMS), Dipartimento di Scienze del Farmaco, Università di Padova, via Marzolo 5, I-35131 Padova, Italy
| | - Stephanie Federico
- Dipartimento di Scienze Chimiche e Farmaceutiche, Università di Trieste, Piazzale Europa 1, I-34127 Trieste, Italy
| | - Giampiero Spalluto
- Dipartimento di Scienze Chimiche e Farmaceutiche, Università di Trieste, Piazzale Europa 1, I-34127 Trieste, Italy
| | - Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810 USA
| |
Collapse
|
38
|
Vernall AJ, Stoddart LA, Briddon SJ, Ng HW, Laughton CA, Doughty SW, Hill SJ, Kellam B. Conversion of a non-selective adenosine receptor antagonist into A3-selective high affinity fluorescent probes using peptide-based linkers. Org Biomol Chem 2013; 11:5673-82. [DOI: 10.1039/c3ob41221k] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|