1
|
Gaitonde SA, Avet C, de la Fuente Revenga M, Blondel-Tepaz E, Shahraki A, Pastor AM, Talagayev V, Robledo P, Kolb P, Selent J, González-Maeso J, Bouvier M. Pharmacological fingerprint of antipsychotic drugs at the serotonin 5-HT 2A receptor. Mol Psychiatry 2024; 29:2753-2764. [PMID: 38561467 PMCID: PMC11420065 DOI: 10.1038/s41380-024-02531-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024]
Abstract
The intricate involvement of the serotonin 5-HT2A receptor (5-HT2AR) both in schizophrenia and in the activity of antipsychotic drugs is widely acknowledged. The currently marketed antipsychotic drugs, although effective in managing the symptoms of schizophrenia to a certain extent, are not without their repertoire of serious side effects. There is a need for better therapeutics to treat schizophrenia for which understanding the mechanism of action of the current antipsychotic drugs is imperative. With bioluminescence resonance energy transfer (BRET) assays, we trace the signaling signature of six antipsychotic drugs belonging to three generations at the 5-HT2AR for the entire spectrum of signaling pathways activated by serotonin (5-HT). The antipsychotic drugs display previously unidentified pathway preference at the level of the individual Gα subunits and β-arrestins. In particular, risperidone, clozapine, olanzapine and haloperidol showed G protein-selective inverse agonist activity. In addition, G protein-selective partial agonism was found for aripiprazole and cariprazine. Pathway-specific apparent dissociation constants determined from functional analyses revealed distinct coupling-modulating capacities of the tested antipsychotics at the different 5-HT-activated pathways. Computational analyses of the pharmacological and structural fingerprints support a mechanistically based clustering that recapitulate the clinical classification (typical/first generation, atypical/second generation, third generation) of the antipsychotic drugs. The study provides a new framework to functionally classify antipsychotics that should represent a useful tool for the identification of better and safer neuropsychiatric drugs and allows formulating hypotheses on the links between specific signaling cascades and in the clinical outcomes of the existing drugs.
Collapse
Affiliation(s)
- Supriya A Gaitonde
- Institute for Research in Immunology and Cancer (IRIC), Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Charlotte Avet
- Institute for Research in Immunology and Cancer (IRIC), Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Mario de la Fuente Revenga
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Elodie Blondel-Tepaz
- Institute for Research in Immunology and Cancer (IRIC), Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Aida Shahraki
- Department of Pharmaceutical Chemistry, Philipps-Universität Marburg, Marbacher Weg 8, 35032, Marburg, Germany
| | - Adrian Morales Pastor
- Research Programme on Biomedical Informatics (GRIB), IMIM-Hospital del Mar Medical Research Institute, Barcelona, 08003, Spain
| | - Valerij Talagayev
- Department of Pharmaceutical Chemistry, Philipps-Universität Marburg, Marbacher Weg 8, 35032, Marburg, Germany
| | - Patricia Robledo
- Integrative Pharmacology and Systems Neuroscience Research Group, IMIM-Hospital del Mar Medical Research Institute, Barcelona, 08003, Spain
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, Philipps-Universität Marburg, Marbacher Weg 8, 35032, Marburg, Germany
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), IMIM-Hospital del Mar Medical Research Institute, Barcelona, 08003, Spain
| | - Javier González-Maeso
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer (IRIC), Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
2
|
Juza R, Musilek K, Mezeiova E, Soukup O, Korabecny J. Recent advances in dopamine D 2 receptor ligands in the treatment of neuropsychiatric disorders. Med Res Rev 2023; 43:55-211. [PMID: 36111795 DOI: 10.1002/med.21923] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 02/04/2023]
Abstract
Dopamine is a biologically active amine synthesized in the central and peripheral nervous system. This biogenic monoamine acts by activating five types of dopamine receptors (D1-5 Rs), which belong to the G protein-coupled receptor family. Antagonists and partial agonists of D2 Rs are used to treat schizophrenia, Parkinson's disease, depression, and anxiety. The typical pharmacophore with high D2 R affinity comprises four main areas, namely aromatic moiety, cyclic amine, central linker and aromatic/heteroaromatic lipophilic fragment. From the literature reviewed herein, we can conclude that 4-(2,3-dichlorophenyl), 4-(2-methoxyphenyl)-, 4-(benzo[b]thiophen-4-yl)-1-substituted piperazine, and 4-(6-fluorobenzo[d]isoxazol-3-yl)piperidine moieties are critical for high D2 R affinity. Four to six atoms chains are optimal for D2 R affinity with 4-butoxyl as the most pronounced one. The bicyclic aromatic/heteroaromatic systems are most frequently occurring as lipophilic appendages to retain high D2 R affinity. In this review, we provide a thorough overview of the therapeutic potential of D2 R modulators in the treatment of the aforementioned disorders. In addition, this review summarizes current knowledge about these diseases, with a focus on the dopaminergic pathway underlying these pathologies. Major attention is paid to the structure, function, and pharmacology of novel D2 R ligands, which have been developed in the last decade (2010-2021), and belong to the 1,4-disubstituted aromatic cyclic amine group. Due to the abundance of data, allosteric D2 R ligands and D2 R modulators from patents are not discussed in this review.
Collapse
Affiliation(s)
- Radomir Juza
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Musilek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Eva Mezeiova
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
3
|
G-protein Biased Signaling Agonists of Dopamine D3 Receptor Promote Distinct Activation Patterns of ERK1/2. Pharmacol Res 2022; 179:106223. [DOI: 10.1016/j.phrs.2022.106223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/31/2022] [Accepted: 04/10/2022] [Indexed: 01/11/2023]
|
4
|
Servant NB, Williams ME, Brust PF, Tang H, Wong MS, Chen Q, Lebl-Rinnova M, Adamski-Werner SL, Tachdjian C, Servant G. A Dynamic Mass Redistribution Assay for the Human Sweet Taste Receptor Uncovers G-Protein Dependent Biased Ligands. Front Pharmacol 2022; 13:832529. [PMID: 35250580 PMCID: PMC8893300 DOI: 10.3389/fphar.2022.832529] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 12/26/2022] Open
Abstract
The sweet taste receptor is rather unique, recognizing a diverse repertoire of natural or synthetic ligands, with a surprisingly large structural diversity, and with potencies stretching over more than six orders of magnitude. Yet, it is not clear if different cell-based assays can faithfully report the relative potencies and efficacies of these molecules. Indeed, up to now, sweet taste receptor agonists have been almost exclusively characterized using cell-based assays developed with overexpressed and promiscuous G proteins. This non-physiological coupling has allowed the quantification of receptor activity via phospholipase C activation and calcium mobilization measurements in heterologous cells on a FLIPR system, for example. Here, we developed a novel assay for the human sweet taste receptor where endogenous G proteins and signaling pathways are recruited by the activated receptor. The effects of several sweet taste receptor agonists and other types of modulators were recorded by measuring changes in dynamic mass redistribution (DMR) using an Epic® reader. Potency and efficacy values obtained in the DMR assay were compared to those results obtained with the classical FLIPR assay. Results demonstrate that for some ligands, the two assay systems provide similar information. However, a clear bias for the FLIPR assay was observed for one third of the agonists evaluated, suggesting that the use of non-physiological coupling may influence the potency and efficacy of sweet taste receptor ligands. Replacing the promiscuous G protein with a chimeric G protein containing the C-terminal tail 25 residues of the physiologically relevant G protein subunit Gαgustducin reduced or abrogated bias.
Collapse
|
5
|
Kolb P, Kenakin T, Alexander SPH, Bermudez M, Bohn LM, Breinholt CS, Bouvier M, Hill SJ, Kostenis E, Martemyanov K, Neubig RR, Onaran HO, Rajagopal S, Roth BL, Selent J, Shukla AK, Sommer ME, Gloriam DE. Community Guidelines for GPCR Ligand Bias: IUPHAR Review XX. Br J Pharmacol 2022; 179:3651-3674. [PMID: 35106752 PMCID: PMC7612872 DOI: 10.1111/bph.15811] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/29/2022] Open
Abstract
G protein-coupled receptors modulate a plethora of physiological processes and mediate the effects of one-third of FDA-approved drugs. Depending on which ligand activates a receptor, it can engage different intracellular transducers. This 'biased signaling' paradigm requires that we now characterize physiological signaling not just by receptors but by ligand-receptor pairs. Ligands eliciting biased signaling may constitute better drugs with higher efficacy and fewer adverse effects. However, ligand bias is very complex, making reproducibility and description challenging. Here, we provide guidelines and terminology for any scientists to design and report ligand bias experiments. The guidelines will aid consistency and clarity, as the basic receptor research and drug discovery communities continue to advance our understanding and exploitation of ligand bias. Scientific insight, biosensors, and analytical methods are still evolving and should benefit from and contribute to the implementation of the guidelines, together improving translation from in vitro to disease-relevant in vivo models.
Collapse
Affiliation(s)
- Peter Kolb
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, North, Carolina, USA
| | | | - Marcel Bermudez
- Department of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Laura M Bohn
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Christian S Breinholt
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Québec, Canada
| | - Stephen J Hill
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Evi Kostenis
- Molecular, Cellular, and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Kirill Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Rick R Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - H Ongun Onaran
- Molecular Biology and Technology Development Unit, Department of Pharmacology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Sudarshan Rajagopal
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.,Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Bryan L Roth
- Department of Pharmacology, University of North Carolina School of Medicine, North, Carolina, USA
| | - Jana Selent
- Research Programme on Biomedical Informatics, Hospital Del Mar Medical Research Institute, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| | - Martha E Sommer
- Institute of Medical Physics and Biophysics, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Current affiliation: ISAR Bioscience Institute, Munich-Planegg, Germany
| | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Plouffe B, Karamitri A, Flock T, Gallion JM, Houston S, Daly CA, Bonnefond A, Guillaume JL, Le Gouill C, Froguel P, Lichtarge O, Deupi X, Jockers R, Bouvier M. Structural Elements Directing G Proteins and β-Arrestin Interactions with the Human Melatonin Type 2 Receptor Revealed by Natural Variants. ACS Pharmacol Transl Sci 2022; 5:89-101. [PMID: 35846981 PMCID: PMC9281605 DOI: 10.1021/acsptsci.1c00239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
G protein-coupled receptors (GPCRs) can engage distinct subsets of signaling pathways, but the structural determinants of this functional selectivity remain elusive. The naturally occurring genetic variants of GPCRs, selectively affecting different pathways, offer an opportunity to explore this phenomenon. We previously identified 40 coding variants of the MTNR1B gene encoding the melatonin MT2 receptor (MT2). These mutations differently impact the β-arrestin 2 recruitment, ERK activation, cAMP production, and Gαi1 and Gαz activation. In this study, we combined functional clustering and structural modeling to delineate the molecular features controlling the MT2 functional selectivity. Using non-negative matrix factorization, we analyzed the signaling signatures of the 40 MT2 variants yielding eight clusters defined by unique signaling features and localized in distinct domains of MT2. Using computational homology modeling, we describe how specific mutations can selectively affect the subsets of signaling pathways and offer a proof of principle that natural variants can be used to explore and understand the GPCR functional selectivity.
Collapse
Affiliation(s)
- Bianca Plouffe
- Department
of Biochemistry and Molecular Medicine, Université de Montréal, H3T 1J4 Montréal, Québec, Canada,Institute
for Research in Immunology and Cancer, Université
de Montréal, H3T 1J4 Montréal, Québec, Canada,The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University Belfast, BT9 7BL Belfast, U.K.
| | - Angeliki Karamitri
- Université
de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Tilman Flock
- Laboratory
of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland,Department
of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Jonathan M. Gallion
- Program
in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, 77030 Houston, Texas, United States
| | - Shane Houston
- The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University Belfast, BT9 7BL Belfast, U.K.
| | - Carole A. Daly
- The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University Belfast, BT9 7BL Belfast, U.K.
| | - Amélie Bonnefond
- Université
de Lille, INSERM/CNRS UMR 1283/8199—EGID, Institut Pasteur
de Lille, CHU de Lille, 59045 Lille, France
| | - Jean-Luc Guillaume
- Université
de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Christian Le Gouill
- Institute
for Research in Immunology and Cancer, Université
de Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Phillipe Froguel
- Université
de Lille, INSERM/CNRS UMR 1283/8199—EGID, Institut Pasteur
de Lille, CHU de Lille, 59045 Lille, France
| | - Olivier Lichtarge
- Program
in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, 77030 Houston, Texas, United States,Department
of Molecular and Human Genetics, Baylor
College of Medicine, 77030 Houston, Texas, United States
| | - Xavier Deupi
- Laboratory
of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland,Condensed
Matter Theory Group, Division of Scientific Computing, Theory, and
Data, Paul Scherrer Institute, 5232 Villigen, Switzerland,. Phone: +41-563103337
| | - Ralf Jockers
- Université
de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France,. Phone: +33-140516434
| | - Michel Bouvier
- Department
of Biochemistry and Molecular Medicine, Université de Montréal, H3T 1J4 Montréal, Québec, Canada,Institute
for Research in Immunology and Cancer, Université
de Montréal, H3T 1J4 Montréal, Québec, Canada,. Phone: 1-514-343-6319
| |
Collapse
|
7
|
Limbird LE. Pushing Forward the Future Tense: Perspectives of a Scientist. Annu Rev Pharmacol Toxicol 2021; 62:1-18. [PMID: 34339291 DOI: 10.1146/annurev-pharmtox-052220-123748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This review is a somewhat chronological tale of my scientific life, emphasizing the why of the questions we asked in the lab and lessons learned that may be of value to nascent scientists. The reader will come to realize that the flow of my life has been driven by a combined life of the mind and life of the soul, intertwining like the strands of DNA. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Lee E Limbird
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee 37208, USA;
| |
Collapse
|
8
|
Onaran HO, Costa T. Conceptual and experimental issues in biased agonism. Cell Signal 2021; 82:109955. [PMID: 33607257 DOI: 10.1016/j.cellsig.2021.109955] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/20/2021] [Accepted: 02/14/2021] [Indexed: 12/31/2022]
Abstract
In this review, we discuss the theoretical and experimental foundations for assessing agonism in the context of signalling bias in GPCRs. We show that the formulation of efficacy in classical receptor theory and the definition of ligand-induced allosteric effect in chemical thermodynamics are coincident measures of agonism, only if we recognize that the classical model cannot be considered as a mechanistic description of the physicochemical events underlying ligand-receptor signalling. It represents instead a mathematical tool, fortuitously capable of extracting efficacy information from concentration-dependent functional data, where both ligand-dependent and ligand-independent information are present. We also assert that dissecting efficacy from affinity, as originally advocated in classical theory, is imperative for understanding the molecular property underlying agonism, and the biased agonism that leads to preferential formation of diverse GPCR-transducer complexes. Finally, we argue that beyond the assumed translational value of functional selectivity (i.e. signalling bias), the identification of ligands with true bias of efficacy is of fundamental importance for unravelling the conformational space that determines the complex functional chemistry of GPCRs.
Collapse
Affiliation(s)
- H Ongun Onaran
- Ankara University, Faculty of Medicine, Department of Pharmacology, Molecular Biology and Technology Development Unit, Ankara, Turkey.
| | | |
Collapse
|
9
|
Bertrand JA, Woodward DF, Sherwood JM, Wang JW, Overby DR. The role of EP 2 receptors in mediating the ultra-long-lasting intraocular pressure reduction by JV-GL1. Br J Ophthalmol 2020; 105:1610-1616. [PMID: 33239414 DOI: 10.1136/bjophthalmol-2020-317762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/23/2020] [Accepted: 10/30/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND A single application of JV-GL1 substantially lowers non-human primate intraocular pressure (IOP) for about a week, independent of dose. This highly protracted effect does not correlate with its ocular biodisposition or correlate with the once-daily dosing regimen for other prostanoid EP2 receptor agonists such as trapenepag or omidenepag. The underlying pharmacological mechanism for the multiday extended activity of JV-GL1 is highly intriguing. The present studies were intended to determine EP2 receptor involvement in mediating the long-term ocular hypotensive activity of JV-GL1 by using mice genetically deficient in EP2 receptors. METHODS The protracted IOP reduction produced by JV-GL1 was investigated in C57BL/6J and EP2 receptor knock-out mice (B6.129-Ptger2tm1Brey /J; EP2KO). Both ocular normotensive and steroid-induced ocular hypertensive (SI-OHT) mice were studied. IOP was measured tonometrically under general anaesthesia. Aqueous humour outflow facility was measured ex vivo using iPerfusion in normotensive C57BL/6J mouse eyes perfused with 100 nM de-esterified JV-GL1 and in SI-OHT C57BL/6J mouse eyes that had received topical JV-GL1 (0.01%) 3 days prior. RESULTS Both the initial 1-day and the protracted multiday effects of JV-GL1 in the SI-OHT model for glaucoma were abolished by deletion of the gene encoding the EP2 receptor. Thus, JV-GL1 did not lower IOP in SI-OHT EP2KO mice, but in littermate SI-OHT EP2WT control mice, JV-GL1 statistically significantly lowered IOP for 4-6 days. CONCLUSIONS Both the 1-day and the long-term effects of JV-GL1 on IOP are entirely EP2 receptor dependent.
Collapse
Affiliation(s)
| | - David F Woodward
- Dept. of Bioengineering, Imperial College London, London, UK.,JeniVision Inc, Suite 200, Irvine, California, USA
| | | | - Jenny W Wang
- JeniVision Inc, Suite 200, Irvine, California, USA
| | - Darryl R Overby
- Dept. of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
10
|
Marti-Solano M, Crilly SE, Malinverni D, Munk C, Harris M, Pearce A, Quon T, Mackenzie AE, Wang X, Peng J, Tobin AB, Ladds G, Milligan G, Gloriam DE, Puthenveedu MA, Babu MM. Combinatorial expression of GPCR isoforms affects signalling and drug responses. Nature 2020; 587:650-656. [PMID: 33149304 PMCID: PMC7611127 DOI: 10.1038/s41586-020-2888-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 09/24/2020] [Indexed: 02/06/2023]
Abstract
G-protein-coupled receptors (GPCRs) are membrane proteins that modulate physiology across human tissues in response to extracellular signals. GPCR-mediated signalling can differ because of changes in the sequence1,2 or expression3 of the receptors, leading to signalling bias when comparing diverse physiological systems4. An underexplored source of such bias is the generation of functionally diverse GPCR isoforms with different patterns of expression across different tissues. Here we integrate data from human tissue-level transcriptomes, GPCR sequences and structures, proteomics, single-cell transcriptomics, population-wide genetic association studies and pharmacological experiments. We show how a single GPCR gene can diversify into several isoforms with distinct signalling properties, and how unique isoform combinations expressed in different tissues can generate distinct signalling states. Depending on their structural changes and expression patterns, some of the detected isoforms may influence cellular responses to drugs and represent new targets for developing drugs with improved tissue selectivity. Our findings highlight the need to move from a canonical to a context-specific view of GPCR signalling that considers how combinatorial expression of isoforms in a particular cell type, tissue or organism collectively influences receptor signalling and drug responses.
Collapse
Affiliation(s)
| | - Stephanie E Crilly
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Duccio Malinverni
- MRC Laboratory of Molecular Biology, Cambridge, UK
- Department of Structural Biology and Center for Data Driven Discovery, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Christian Munk
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Matthew Harris
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Abigail Pearce
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Tezz Quon
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Amanda E Mackenzie
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Biology, University of North Dakota, Grand Forks, ND, USA
| | - Junmin Peng
- Center for Proteomics and Metabolomics, St Jude Children's Research Hospital, Memphis, TN, USA
- Departments of Structural Biology and Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Andrew B Tobin
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Manojkumar A Puthenveedu
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - M Madan Babu
- MRC Laboratory of Molecular Biology, Cambridge, UK.
- Department of Structural Biology and Center for Data Driven Discovery, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
11
|
Scarpa M, Hesse S, Bradley SJ. M1 muscarinic acetylcholine receptors: A therapeutic strategy for symptomatic and disease-modifying effects in Alzheimer's disease? ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 88:277-310. [PMID: 32416870 DOI: 10.1016/bs.apha.2019.12.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The M1 muscarinic acetylcholine receptor (mAChR) plays a crucial role in learning and memory processes and has long been identified as a promising therapeutic target for the improvement of cognitive decline in Alzheimer's disease (AD). As such, clinical trials with xanomeline, a mAChR orthosteric agonist, showed an improvement in cognitive and behavioral symptoms associated with AD. Despite this, the clinical utility of xanomeline was hampered by a lack of M1 receptor selectivity and adverse cholinergic responses attributed to activation of peripheral M2 and M3 mAChRs. More recently, efforts have focused on developing more selective M1 compounds via targeting the less-conserved allosteric binding pockets. As such, positive allosteric modulators (PAMs) have emerged as an exciting strategy to achieve exquisite selectivity for the M1 mAChR in order to deliver improvements in cognitive function in AD. Furthermore, over recent years it has become increasingly apparent that M1 therapeutics may also offer disease-modifying effects in AD, due to the modulation of pathogenic amyloid processing. This article will review the progress made in the development of M1 selective ligands for the treatment of cognitive decline in AD, and will discuss the current evidence that selective targeting of the M1 mAChR could also have the potential to modify AD progression.
Collapse
Affiliation(s)
- Miriam Scarpa
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sarah Hesse
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sophie J Bradley
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
12
|
De Vries L, Finana F, Cathala C, Ronsin B, Cussac D. Innovative Bioluminescence Resonance Energy Transfer Assay Reveals Differential Agonist-Induced D2 Receptor Intracellular Trafficking and Arrestin-3 Recruitment. Mol Pharmacol 2019; 96:308-319. [PMID: 31266815 DOI: 10.1124/mol.119.115998] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/19/2019] [Indexed: 01/14/2023] Open
Abstract
The dopamine D2 receptor (D2R) mediates ligand-biased signaling with potential therapeutic implications. However, internalization, choice of endocytic routes, and degradation of the D2R in lysosomes may also participate in agonist-directed trafficking. We developed bioluminescence resonance energy transfer (BRET) assays that measure relative distances between Renilla luciferase8-tagged D2R and green fluorescent protein 2 (GFP2)-tagged K-Ras (plasma membrane marker), and between luciferase8-tagged D2R and GFP2-Rab5 (early), GFP2-Rab4 (recycling), or GFP2-Rab7 (late) endosomal markers. The BRET signal between D2R-Luc and GFP2-K-Ras was robustly diminished after receptor internalization induced by dopamine, with subsequent BRET signals increasing when luciferase8-tagged D2R approached GFP2-Rab proteins in endosomal compartments. All BRET signals were blocked by the selective D2R antagonist haloperidol and were decreased by low temperature and high sucrose blocks, two parameters interfering with internalization. Some antipsychotic drugs, such as aripiprazole, are less efficacious in internalizing D2R than most of the antiparkinsonian agents. However, antipsychotics were nearly as efficacious as antiparkinsonians in directing the D2R toward early and recycling endosomes. The Rab7 marker for the late endosome/lysosome route was also capable of discriminating between D2R compounds. We could show that some drugs engaged the D2R either to interact preferentially with arrestin-3 or to internalize. Our study revealed that D2R trafficking in cells was differentially regulated by antipsychotic and antiparkinsonian drugs. Taken together, the BRET assays reported here could further help decipher D2R ligand-induced arrestin-3 recruitment and trafficking, with potentially more selective therapeutic profiles and fewer undesired side effects.
Collapse
Affiliation(s)
- Luc De Vries
- Central Nervous System Innovation Unit, CEPC Campans - Belair de Campans, Castres, France (L.D.V., F.F., C.C., D.C.) and CNRS, UMR5547, Centre de Biologie du Développement, Université de Toulouse III-Paul Sabatier, Toulouse, France (B.R.)
| | - Frédéric Finana
- Central Nervous System Innovation Unit, CEPC Campans - Belair de Campans, Castres, France (L.D.V., F.F., C.C., D.C.) and CNRS, UMR5547, Centre de Biologie du Développement, Université de Toulouse III-Paul Sabatier, Toulouse, France (B.R.)
| | - Claudie Cathala
- Central Nervous System Innovation Unit, CEPC Campans - Belair de Campans, Castres, France (L.D.V., F.F., C.C., D.C.) and CNRS, UMR5547, Centre de Biologie du Développement, Université de Toulouse III-Paul Sabatier, Toulouse, France (B.R.)
| | - Brice Ronsin
- Central Nervous System Innovation Unit, CEPC Campans - Belair de Campans, Castres, France (L.D.V., F.F., C.C., D.C.) and CNRS, UMR5547, Centre de Biologie du Développement, Université de Toulouse III-Paul Sabatier, Toulouse, France (B.R.)
| | - Didier Cussac
- Central Nervous System Innovation Unit, CEPC Campans - Belair de Campans, Castres, France (L.D.V., F.F., C.C., D.C.) and CNRS, UMR5547, Centre de Biologie du Développement, Université de Toulouse III-Paul Sabatier, Toulouse, France (B.R.)
| |
Collapse
|
13
|
Affiliation(s)
- Sophie G Martin
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
14
|
Wang J, Miao Y. Mechanistic Insights into Specific G Protein Interactions with Adenosine Receptors. J Phys Chem B 2019; 123:6462-6473. [PMID: 31283874 DOI: 10.1021/acs.jpcb.9b04867] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Coupling between G-protein-coupled receptors (GPCRs) and the G proteins is a key step in cellular signaling. Despite extensive experimental and computational studies, the mechanism of specific GPCR-G protein coupling remains poorly understood. This has greatly hindered effective drug design of GPCRs that are primary targets of ∼1/3 of currently marketed drugs. Here, we have employed all-atom simulations using a robust Gaussian accelerated molecular dynamics (GaMD) method to decipher the mechanism of the GPCR-G protein interactions. Adenosine receptors (ARs) were used as model systems based on very recently determined cryo-EM structures of the A1AR and A2AAR coupled with the Gi and Gs proteins, respectively. Changing the Gi protein to the Gs led to increased fluctuations in the A1AR and agonist adenosine (ADO), while agonist 5'-N-ethylcarboxamidoadenosine (NECA) binding in the A2AAR could be still stabilized upon changing the Gs protein to the Gi. Free energy calculations identified one stable low-energy conformation for each of the A1AR-Gi and A2AAR-Gs complexes as in the cryo-EM structures, similarly for the A2AAR-Gi complex. In contrast, the ADO agonist and Gs protein sampled multiple conformations in the A1AR-Gs system. GaMD simulations thus indicated that the A1AR preferred to couple with the Gi protein to the Gs, while the A2AAR could couple with both the Gs and Gi proteins, being highly consistent with experimental findings of the ARs. More importantly, detailed analysis of the atomic simulations showed that the specific AR-G protein coupling resulted from remarkably complementary residue interactions at the protein interface, involving mainly the receptor transmembrane 6 helix and the Gα α5 helix and α4-β6 loop. In summary, the GaMD simulations have provided unprecedented insights into the dynamic mechanism of specific GPCR-G protein interactions at an atomistic level.
Collapse
Affiliation(s)
- Jinan Wang
- Center for Computational Biology and Department of Molecular Biosciences , University of Kansas , Lawrence , Kansas 66047 , United States
| | - Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences , University of Kansas , Lawrence , Kansas 66047 , United States
| |
Collapse
|
15
|
Mahmod Al-Qattan MN, Mordi MN. Molecular Basis of Modulating Adenosine Receptors Activities. Curr Pharm Des 2019; 25:817-831. [DOI: 10.2174/1381612825666190304122624] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 02/26/2019] [Indexed: 01/04/2023]
Abstract
Modulating cellular processes through extracellular chemical stimuli is medicinally an attractive approach to control disease conditions. GPCRs are the most important group of transmembranal receptors that produce different patterns of activations using intracellular mediators (such as G-proteins and Beta-arrestins). Adenosine receptors (ARs) belong to GPCR class and are divided into A1AR, A2AAR, A2BAR and A3AR. ARs control different physiological activities thus considered valuable target to control neural, heart, inflammatory and other metabolic disorders. Targeting ARs using small molecules essentially works by binding orthosteric and/or allosteric sites of the receptors. Although targeting orthosteric site is considered typical to modulate receptor activity, allosteric sites provide better subtype selectivity, saturable modulation of activity and variable activation patterns. Each receptor exists in dynamical equilibrium between conformational ensembles. The equilibrium is affected by receptor interaction with other molecules. Changing the population of conformational ensembles of the receptor is the method by which orthosteric, allosteric and other cellular components control receptor signaling. Herein, the interactions of ARs with orthosteric, allosteric ligands as well as intracellular mediators are described. A quinary interaction model for the receptor is proposed and energy wells for major conformational ensembles are retrieved.
Collapse
Affiliation(s)
| | - Mohd Nizam Mordi
- Centre For Drug Research, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia
| |
Collapse
|
16
|
Characterisation of small molecule ligands 4CMTB and 2CTAP as modulators of human FFA2 receptor signalling. Sci Rep 2018; 8:17819. [PMID: 30546040 PMCID: PMC6292860 DOI: 10.1038/s41598-018-36242-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/30/2018] [Indexed: 01/22/2023] Open
Abstract
Short chain fatty acids (SCFAs) are protective against inflammatory diseases. Free fatty acid receptor 2 (FFA2), is a target of SCFAs however, their selectivity for FFA2 over other FFA receptors is limited. This study aimed to functionally characterise 2-(4-chlorophenyl)-3-methyl-N-(thiazole-2-yl)butanamide (4CMTB) and 4-((4-(2-chlorophenyl)thiazole-2-yl)amino)-4oxo-3-phenylbutanoic acid (2CTAP), and their enantiomers, in modulating FFA2 activity. The racemic mixture (R/S) and its constituents (R-) and (S-) 4CMTB or 2CTAP were used to stimulate human (h)FFA2 in the absence or presence of acetate. Calcium ions (Ca2+), phosphorylated extracellular signal-regulated kinase 1 and 2 (pERK1/2) and cyclic adenosine monophosphate (cAMP) were measured. R/S-4CMTB is a functionally selective ago-allosteric ligand that enhances Ca2+ response to acetate. Both R/S-4CMTB and S-4CMTB are more potent activators of pERK1/2 and inhibitors of forskolin-induced cAMP than acetate. S-4CMTB increased neutrophil infiltration in intestinal ischemia reperfusion injury (IRI). 2CTAP inhibited constitutive Ca2+ levels, antagonised acetate-induced pERK1/2 and prevented damage following IRI. This study characterises enantiomers of functionally selective ligands for FFA2 in cells stably expressing hFFA2. It highlights the novel roles of selective FFA2 enantiomers 4CMTB and 2CTAP on Ca2+, pERK1/2 and cAMP and their roles as allosteric modulators which, may assist in efforts to design novel therapeutic agents for FFA2-driven inflammatory diseases.
Collapse
|
17
|
Ramirez VT, van Oeffelen WEPA, Torres-Fuentes C, Chruścicka B, Druelle C, Golubeva AV, van de Wouw M, Dinan TG, Cryan JF, Schellekens H. Differential functional selectivity and downstream signaling bias of ghrelin receptor antagonists and inverse agonists. FASEB J 2018; 33:518-531. [PMID: 30020830 DOI: 10.1096/fj.201800655r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The ghrelin receptor [growth hormone secretagogue receptor (GHSR)-1a] represents a promising pharmacologic target for the treatment of metabolic disorders, including obesity and cachexia, via central appetite modulation. The GHSR-1a has a complex pharmacology, highlighted by G-protein-dependent and -independent downstream signaling pathways and high basal constitutive activity. The functional selectivity and signaling bias of many GHSR-1a-specific ligands has not been fully characterized. In this study, we investigated the pharmacologic properties of ghrelin, MK-0677, L692,585, and [d-Lys3]-growth hormone-releasing peptide-6 (Dlys), JMV2959, and [d-Arg(1),d-Phe(5),d-Trp(7, 9),Leu(11)]-substance P (SP-analog). We investigated their effect on basal GHSR-1a constitutive signaling, ligand-directed downstream GHSR-1a signaling, functional selectivity, and signaling bias. Dlys behaved as a partial antagonist with a strong bias toward GHSR-1a-β-arrestin signaling, whereas JMV2959 acted as a full unbiased GHSR-1a antagonist. Moreover, the SP-analog behaved as an inverse agonist increasing G-protein-dependent signaling, but only at high concentrations, whereas, at low concentrations, the SP-analog attenuated β-arrestin-dependent signaling. Considering the limited success in the clinical development of GHSR-1a-targeted drugs so far, these findings provide a novel insight into the pharmacologic characteristics of GHSR-1a ligands and their signaling bias, which has important implications in the design of novel, more selective GHSR-1a ligands with predictable functional outcome and selectivity for preclinical and clinical drug development.-Ramirez, V. T., van Oeffelen, W. E. P. A., Torres-Fuentes, C., Chruścicka, B., Druelle, C., Golubeva, A. V., van de Wouw, M., Dinan, T. G., Cryan, J. F., Schellekens, H. Differential functional selectivity and downstream signaling bias of ghrelin receptor antagonists and inverse agonists.
Collapse
Affiliation(s)
- Valerie T Ramirez
- Alimentary Pharmabiotic Centre (APC) Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Cristina Torres-Fuentes
- Alimentary Pharmabiotic Centre (APC) Microbiome Ireland, University College Cork, Cork, Ireland
| | - Barbara Chruścicka
- Alimentary Pharmabiotic Centre (APC) Microbiome Ireland, University College Cork, Cork, Ireland
| | - Clementine Druelle
- Alimentary Pharmabiotic Centre (APC) Microbiome Ireland, University College Cork, Cork, Ireland
| | - Anna V Golubeva
- Alimentary Pharmabiotic Centre (APC) Microbiome Ireland, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- Alimentary Pharmabiotic Centre (APC) Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- Alimentary Pharmabiotic Centre (APC) Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry, University College Cork, Cork, Ireland; and
| | - John F Cryan
- Alimentary Pharmabiotic Centre (APC) Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,Food for Health Ireland, University College Cork, Cork, Ireland
| | - Harriët Schellekens
- Alimentary Pharmabiotic Centre (APC) Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,Food for Health Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
18
|
Bradley SJ, Tobin AB, Prihandoko R. The use of chemogenetic approaches to study the physiological roles of muscarinic acetylcholine receptors in the central nervous system. Neuropharmacology 2018; 136:421-426. [PMID: 29191752 DOI: 10.1016/j.neuropharm.2017.11.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/12/2017] [Accepted: 11/26/2017] [Indexed: 12/19/2022]
Abstract
Chemical genetic has played an important role in linking specific G protein-coupled receptor (GPCR) signalling to cellular processes involved in central nervous system (CNS) functions. Key to this approach has been the modification of receptor properties such that receptors no longer respond to endogenous ligands but rather can be activated selectively by synthetic ligands. Such modified receptors have been called Receptors Activated Solely by Synthetic Ligands (RASSLs) or Designer Receptors Exclusively Activated by Designer Drugs (DREADDs). Unlike knock-out animal models which allow detection of phenotypic changes caused by loss of receptor functions, RASSL and DREADD receptors offer the possibility of rescuing "knock-out" phenotypic deficits by administration of the synthetic ligands. Here we describe the use of these modified receptors in defining the physiological role of GPCRs and validation of receptors as drug targets. This article is part of the Special Issue entitled 'Neuropharmacology on Muscarinic Receptors'.
Collapse
Affiliation(s)
- Sophie J Bradley
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Andrew B Tobin
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Rudi Prihandoko
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| |
Collapse
|
19
|
Hanyaloglu AC. Advances in Membrane Trafficking and Endosomal Signaling of G Protein-Coupled Receptors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 339:93-131. [PMID: 29776606 DOI: 10.1016/bs.ircmb.2018.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The integration of GPCR signaling with membrane trafficking, as a single orchestrated system, is a theme increasingly evident with the growing reports of GPCR endosomal signaling. Once viewed as a mechanism to regulate cell surface heterotrimeric G protein signaling, the endocytic trafficking system is complex, highly compartmentalized, yet deeply interconnected with cell signaling. The organization of receptors into distinct plasma membrane signalosomes, biochemically distinct endosomal populations, endosomal microdomains, and its communication with distinct subcellular organelles such as the Golgi provides multiple unique signaling platforms that are critical for specifying receptor function physiologically and pathophysiologically. In this chapter I discuss our emerging understanding in the endocytic trafficking systems employed by GPCRs and their novel roles in spatial control of signaling. Given the extensive roles that GPCRs play in vivo, these evolving models are starting to provide mechanistic understanding of distinct diseases and provide novel therapeutic avenues that are proving to be viable targets.
Collapse
Affiliation(s)
- Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, London, United Kingdom.
| |
Collapse
|
20
|
Evolving View of Membrane Trafficking and Signaling Systems for G Protein-Coupled Receptors. ENDOCYTOSIS AND SIGNALING 2018; 57:273-299. [DOI: 10.1007/978-3-319-96704-2_10] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
Gabl M, Holdfeldt A, Sundqvist M, Lomei J, Dahlgren C, Forsman H. FPR2 signaling without β-arrestin recruitment alters the functional repertoire of neutrophils. Biochem Pharmacol 2017; 145:114-122. [DOI: 10.1016/j.bcp.2017.08.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 08/23/2017] [Indexed: 01/01/2023]
|
22
|
Spatial encryption of G protein-coupled receptor signaling in endosomes; Mechanisms and applications. Biochem Pharmacol 2017; 143:1-9. [DOI: 10.1016/j.bcp.2017.04.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/25/2017] [Indexed: 01/14/2023]
|
23
|
Systematic errors in detecting biased agonism: Analysis of current methods and development of a new model-free approach. Sci Rep 2017; 7:44247. [PMID: 28290478 PMCID: PMC5349545 DOI: 10.1038/srep44247] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/06/2017] [Indexed: 11/08/2022] Open
Abstract
Discovering biased agonists requires a method that can reliably distinguish the bias in signalling due to unbalanced activation of diverse transduction proteins from that of differential amplification inherent to the system being studied, which invariably results from the non-linear nature of biological signalling networks and their measurement. We have systematically compared the performance of seven methods of bias diagnostics, all of which are based on the analysis of concentration-response curves of ligands according to classical receptor theory. We computed bias factors for a number of β-adrenergic agonists by comparing BRET assays of receptor-transducer interactions with Gs, Gi and arrestin. Using the same ligands, we also compared responses at signalling steps originated from the same receptor-transducer interaction, among which no biased efficacy is theoretically possible. In either case, we found a high level of false positive results and a general lack of correlation among methods. Altogether this analysis shows that all tested methods, including some of the most widely used in the literature, fail to distinguish true ligand bias from "system bias" with confidence. We also propose two novel semi quantitative methods of bias diagnostics that appear to be more robust and reliable than currently available strategies.
Collapse
|
24
|
Costa-Neto CM, Parreiras-E-Silva LT, Bouvier M. A Pluridimensional View of Biased Agonism. Mol Pharmacol 2016; 90:587-595. [PMID: 27638872 DOI: 10.1124/mol.116.105940] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 09/14/2016] [Indexed: 12/17/2022] Open
Abstract
When studying G protein-coupled receptor (GPCR) signaling and ligand-biased agonism, at least three dimensional spaces must be considered, as follows: 1) the distinct conformations that can be stabilized by different ligands promoting the engagement of different signaling effectors and accessory regulators; 2) the distinct subcellular trafficking that can be conferred by different ligands, which results in spatially distinct signals; and 3) the differential binding kinetics that maintain the receptor in specific conformation and/or subcellular localization for different periods of time, allowing for the engagement of distinct signaling effector subsets. These three pluridimensional aspects of signaling contribute to different faces of functional selectivity and provide a complex, interconnected way to define the signaling profile of each individual ligand acting at GPCRs. In this review, we discuss how each of these aspects may contribute to the diversity of signaling, but also how they shed light on the complexity of data analyses and interpretation. The impact of phenotype variability as a source of signaling diversity, and the influence of novel and more sensitive assays in the detection and analysis of signaling pluridimensionality, is also discussed. Finally, we discuss perspectives for the use of the concept of pluridimensional signaling in drug discovery, in which we highlight future predictive tools that may facilitate the identification of compounds with optimal therapeutic and safety properties based on the signaling signatures of drug candidates.
Collapse
Affiliation(s)
- Claudio M Costa-Neto
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil (C.M.C.-N., L.T.P.-S.); and Department of Biochemistry and Molecular Medicine and Institute for Research in Immunology and Cancer, University of Montréal, Montréal, Canada (L.T.P.-S., M.B.)
| | - Lucas T Parreiras-E-Silva
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil (C.M.C.-N., L.T.P.-S.); and Department of Biochemistry and Molecular Medicine and Institute for Research in Immunology and Cancer, University of Montréal, Montréal, Canada (L.T.P.-S., M.B.)
| | - Michel Bouvier
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil (C.M.C.-N., L.T.P.-S.); and Department of Biochemistry and Molecular Medicine and Institute for Research in Immunology and Cancer, University of Montréal, Montréal, Canada (L.T.P.-S., M.B.)
| |
Collapse
|
25
|
Lieb S, Littmann T, Plank N, Felixberger J, Tanaka M, Schäfer T, Krief S, Elz S, Friedland K, Bernhardt G, Wegener J, Ozawa T, Buschauer A. Label-free versus conventional cellular assays: Functional investigations on the human histamine H 1 receptor. Pharmacol Res 2016; 114:13-26. [PMID: 27751876 DOI: 10.1016/j.phrs.2016.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 10/11/2016] [Accepted: 10/11/2016] [Indexed: 01/07/2023]
Abstract
A set of histamine H1 receptor (H1R) agonists and antagonists was characterized in functional assays, using dynamic mass redistribution (DMR), electric cell-substrate impedance sensing (ECIS) and various signaling pathway specific readouts (Fura-2 and aequorin calcium assays, arrestin recruitment (luciferase fragment complementation) assay, luciferase gene reporter assay). Data were gained from genetically engineered HEK293T cells and compared with reference data from GTPase assays and radioligand binding. Histamine and the other H1R agonists gave different assay-related pEC50 values, however, the order of potency was maintained. In the luciferase fragment complementation assay, the H1R preferred β-arrestin2 over β-arrestin1. The calcium and the impedimetric assay depended on Gq coupling of the H1R, as demonstrated by complete inhibition of the histamine-induced signals in the presence of the Gq inhibitor FR900359 (UBO-QIC). Whereas partial inhibition by FR900359 was observed in DMR and the gene reporter assay, pertussis toxin substantially decreased the response in DMR, but increased the luciferase signal, reflecting the contribution of both, Gq and Gi, to signaling in these assays. For antagonists, the results from DMR were essentially compatible with those from conventional readouts, whereas the impedance-based data revealed a trend towards higher pKb values. ECIS and calcium assays apparently only reflect Gq signaling, whereas DMR and gene reporter assays appear to integrate both, Gq and Gi mediated signaling. The results confirm the value of the label-free methods, DMR and ECIS, for the characterization of H1R ligands. Both noninvasive techniques are complementary to each other, but cannot fully replace reductionist signaling pathway focused assays.
Collapse
Affiliation(s)
- S Lieb
- Institute of Pharmacy, University of Regensburg, D-93040 Regensburg, Germany
| | - T Littmann
- Institute of Pharmacy, University of Regensburg, D-93040 Regensburg, Germany
| | - N Plank
- Institute of Pharmacy, University of Regensburg, D-93040 Regensburg, Germany
| | - J Felixberger
- Institute of Pharmacy, University of Regensburg, D-93040 Regensburg, Germany
| | - M Tanaka
- Department of Chemistry, School of Science, University of Tokyo, Tokyo, Japan
| | - T Schäfer
- Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, D-91058 Erlangen, Germany
| | - S Krief
- Bioprojet Biotech, 35762 Saint-Grégoire, France
| | - S Elz
- Institute of Pharmacy, University of Regensburg, D-93040 Regensburg, Germany
| | - K Friedland
- Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, D-91058 Erlangen, Germany
| | - G Bernhardt
- Institute of Pharmacy, University of Regensburg, D-93040 Regensburg, Germany
| | - J Wegener
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, D-93040 Regensburg, Germany
| | - T Ozawa
- Department of Chemistry, School of Science, University of Tokyo, Tokyo, Japan
| | - A Buschauer
- Institute of Pharmacy, University of Regensburg, D-93040 Regensburg, Germany.
| |
Collapse
|
26
|
Multivalent approaches and beyond: novel tools for the investigation of dopamine D2 receptor pharmacology. Future Med Chem 2016; 8:1349-72. [DOI: 10.4155/fmc-2016-0010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The dopamine D2 receptor (D2R) has been implicated in the symptomology of disorders such as schizophrenia and Parkinson's disease. Multivalent ligands provide useful tools to investigate emerging concepts of G protein-coupled receptor drug action such as allostery, bitopic binding and receptor dimerization. This review focuses on the approaches taken toward the development of multivalent ligands for the D2R recently and highlights the challenges associated with each approach, their utility in probing D2R function and approaches to develop new D2R-targeting drugs. Furthermore, we extend our discussion to the possibility of designing multitarget ligands. The insights gained from such studies may provide the basis for improved therapeutic targeting of the D2R.
Collapse
|
27
|
Milanos L, Brox R, Frank T, Poklukar G, Palmisano R, Waibel R, Einsiedel J, Dürr M, Ivanović-Burmazović I, Larsen O, Hjortø GM, Rosenkilde MM, Tschammer N. Discovery and Characterization of Biased Allosteric Agonists of the Chemokine Receptor CXCR3. J Med Chem 2016; 59:2222-43. [DOI: 10.1021/acs.jmedchem.5b01965] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Lampros Milanos
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Regine Brox
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Theresa Frank
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Gašper Poklukar
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Ralf Palmisano
- Optical
Imaging Center Erlangen, Friedrich Alexander University, Hartmannstraße
14, 91052 Erlangen, Germany
| | - Reiner Waibel
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Jürgen Einsiedel
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Maximilian Dürr
- Department
of Chemistry and Pharmacy, Bioorganic Chemistry, Friedrich Alexander University, Egerlandstraße 1, 91058 Erlangen, Germany
| | - Ivana Ivanović-Burmazović
- Department
of Chemistry and Pharmacy, Bioorganic Chemistry, Friedrich Alexander University, Egerlandstraße 1, 91058 Erlangen, Germany
| | - Olav Larsen
- Department
of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology,
Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| | - Gertrud Malene Hjortø
- Department
of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology,
Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| | - Mette Marie Rosenkilde
- Department
of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology,
Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| | - Nuska Tschammer
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| |
Collapse
|
28
|
|
29
|
Quantification of adenosine A 1 receptor biased agonism: Implications for drug discovery. Biochem Pharmacol 2016; 99:101-12. [DOI: 10.1016/j.bcp.2015.11.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/09/2015] [Indexed: 12/20/2022]
|
30
|
In vivo veritas, the next frontier for functionally selective GPCR ligands. Methods 2015; 92:64-71. [PMID: 26320830 DOI: 10.1016/j.ymeth.2015.08.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/22/2015] [Accepted: 08/24/2015] [Indexed: 01/11/2023] Open
Abstract
The realization that G-protein coupled receptors (GPCR) engage several cell signaling mechanisms simultaneously has led to a multiplication of research aimed at developing biased ligands exerting a selective action on subsets of responses downstream of a given receptor. Several tools have been developed to identify such ligands using recombinant cell systems. However the validation of biased ligand activity in animal models remains a serious challenge. Here we present a general strategy that can be used to validate biased ligand activity in vivo and supports it as a strategy for further drug development. In doing so, we placed special attention on strategies allowing to discriminate between G-protein and beta-arrestin mediated mechanisms. We also underscore differences between in vitro and in vivo systems and suggest avenues for tool development to streamline the translation of biased ligands development to pre-clinical animal models.
Collapse
|
31
|
Stroth N, Niso M, Colabufo NA, Perrone R, Svenningsson P, Lacivita E, Leopoldo M. Arylpiperazine agonists of the serotonin 5-HT1A receptor preferentially activate cAMP signaling versus recruitment of β-arrestin-2. Bioorg Med Chem 2015; 23:4824-4830. [PMID: 26081758 DOI: 10.1016/j.bmc.2015.05.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 10/23/2022]
Abstract
G protein-coupled receptors (GPCRs) mediate biological signal transduction through complex molecular pathways. Therapeutic effects of GPCR-directed drugs are typically accompanied by unwanted side effects, owing in part to the parallel engagement of multiple signaling mechanisms. The discovery of drugs that are 'functionally selective' towards therapeutic effects, based on their selective control of cellular responses through a given GPCR, is thus a major goal in pharmacology today. In the present study, we show that several arylpiperazine ligands of the serotonin 5-HT1A receptor (5-HT1AR) preferentially activate 3',5'-cyclic adenosine monophosphate (cAMP) signaling versus β-arrestin-2 recruitment. The pharmacology of these compounds is thus qualitatively different from the endogenous agonist serotonin, indicating functional selectivity of 5-HT1AR-mediated response pathways. Preliminary evidence suggests that phosphorylation of extracellular signal-regulated kinases 1/2 (ERK1/2) downstream of 5-HT1AR is a substrate of functionally selective signaling by partial agonists. We propose that the compounds described in the present study are useful starting points for the development of signaling pathway-selective drugs targeting 5-HT1AR.
Collapse
Affiliation(s)
- Nikolas Stroth
- Center for Molecular Medicine, Department of Neurology and Clinical Neuroscience, Karolinska Institute and Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Mauro Niso
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari 'A. Moro', via Orabona, 4, 70125 Bari, Italy
| | - Nicola A Colabufo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari 'A. Moro', via Orabona, 4, 70125 Bari, Italy
| | - Roberto Perrone
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari 'A. Moro', via Orabona, 4, 70125 Bari, Italy
| | - Per Svenningsson
- Center for Molecular Medicine, Department of Neurology and Clinical Neuroscience, Karolinska Institute and Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Enza Lacivita
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari 'A. Moro', via Orabona, 4, 70125 Bari, Italy
| | - Marcello Leopoldo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari 'A. Moro', via Orabona, 4, 70125 Bari, Italy.
| |
Collapse
|
32
|
Schulte G. Frizzleds and WNT/β-catenin signaling--The black box of ligand-receptor selectivity, complex stoichiometry and activation kinetics. Eur J Pharmacol 2015; 763:191-5. [PMID: 26003275 DOI: 10.1016/j.ejphar.2015.05.031] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/13/2015] [Indexed: 10/23/2022]
Abstract
The lipoglycoproteins of the mammalian WNT family induce β-catenin-dependent signaling through interaction with members of the Class Frizzled receptors and LDL receptor-related protein 5/6 (LRP5/6) albeit with unknown selectivity. The 10 mammalian Frizzleds (FZDs) are seven transmembrane (7TM) spanning receptors and have recently been classified as G protein-coupled receptors (GPCRs). This review summarizes the current knowledge about WNT/FZD selectivity and functional selectivity, the role of co-receptors for signal specification, the formation of receptor complexes as well as the kinetics and mechanisms of signal initiation with focus on WNT/β-catenin signaling. In order to exploit the true therapeutic potential of WNT/FZD signaling to treat human disease, it is clear that substantial progress in the understanding of receptor complex formation and signal specification has to precede a mechanism-based drug design targeting WNT receptors.
Collapse
Affiliation(s)
- Gunnar Schulte
- Department of Physiology & Pharmacology, Section of Receptor Biology & Signaling, Karolinska Institutet, S-17177 Stockholm, Sweden; Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
33
|
Walther C, Ferguson SSG. Minireview: Role of intracellular scaffolding proteins in the regulation of endocrine G protein-coupled receptor signaling. Mol Endocrinol 2015; 29:814-30. [PMID: 25942107 DOI: 10.1210/me.2015-1091] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The majority of hormones stimulates and mediates their signal transduction via G protein-coupled receptors (GPCRs). The signal is transmitted into the cell due to the association of the GPCRs with heterotrimeric G proteins, which in turn activates an extensive array of signaling pathways to regulate cell physiology. However, GPCRs also function as scaffolds for the recruitment of a variety of cytoplasmic protein-interacting proteins that bind to both the intracellular face and protein interaction motifs encoded by GPCRs. The structural scaffolding of these proteins allows GPCRs to recruit large functional complexes that serve to modulate both G protein-dependent and -independent cellular signaling pathways and modulate GPCR intracellular trafficking. This review focuses on GPCR interacting PSD95-disc large-zona occludens domain containing scaffolds in the regulation of endocrine receptor signaling as well as their potential role as therapeutic targets for the treatment of endocrinopathies.
Collapse
Affiliation(s)
- Cornelia Walther
- J. Allyn Taylor Centre for Cell Biology (C.W., S.S.G.F.), Robarts Research Institute, and Department of Physiology and Pharmacology (S.S.G.F.), University of Western Ontario, London, Ontario, Canada N6A 5K8
| | - Stephen S G Ferguson
- J. Allyn Taylor Centre for Cell Biology (C.W., S.S.G.F.), Robarts Research Institute, and Department of Physiology and Pharmacology (S.S.G.F.), University of Western Ontario, London, Ontario, Canada N6A 5K8
| |
Collapse
|
34
|
Christopoulos A. Advances in G protein-coupled receptor allostery: from function to structure. Mol Pharmacol 2014; 86:463-78. [PMID: 25061106 DOI: 10.1124/mol.114.094342] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It is now widely accepted that G protein-coupled receptors (GPCRs) are highly dynamic proteins that adopt multiple active states linked to distinct functional outcomes. Furthermore, these states can be differentially stabilized not only by orthosteric ligands but also by allosteric ligands acting at spatially distinct binding sites. The key pharmacologic characteristics of GPCR allostery include improved selectivity due to either greater sequence divergence between receptor subtypes and/or subtype-selective cooperativity, a ceiling level to the effect, probe dependence (whereby the magnitude and direction of the allosteric effect change with the nature of the interacting ligands), and the potential for biased signaling. Recent chemical biology developments are beginning to demonstrate how the incorporation of analytical pharmacology and operational modeling into the experimental workflow can enrich structure-activity studies of allostery and bias, and have also led to the discovery of a new class of hybrid orthosteric/allosteric (bitopic) molecules. The potential for endogenous allosteric modulators to play a role in physiology and disease remains to be fully appreciated but will likely represent an important area for future studies. Finally, breakthroughs in structural and computational biology are beginning to unravel the mechanistic basis of GPCR allosteric modulation at the molecular level.
Collapse
Affiliation(s)
- Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
35
|
Kelly E. Efficacy and ligand bias at the μ-opioid receptor. Br J Pharmacol 2014; 169:1430-46. [PMID: 23646826 DOI: 10.1111/bph.12222] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 04/10/2013] [Accepted: 04/20/2013] [Indexed: 12/11/2022] Open
Abstract
In order to describe drug action at a GPCR, a full understanding of the pharmacological terms affinity, efficacy and potency is necessary. This is true whether comparing the ability of different agonists to produce a measurable response in a cell or tissue, or determining the relative ability of an agonist to activate a single receptor subtype and produce multiple responses. There is a great deal of interest in the μ-opioid receptor (MOP receptor) and the ligands that act at this GPCR not only because of the clinically important analgesic effects produced by MOP agonists but also because of their liability to induce adverse effects such as respiratory depression and dependence. Our understanding of the mechanisms underlying these effects, as well as the ability to develop new, more effective MOP receptor drugs, depends upon the accurate determination of the efficacy with which these ligands induce coupling of MOP receptors to downstream signalling events. In this review, which is written with the minimum of mathematical content, the basic meaning of terms including efficacy, intrinsic activity and intrinsic efficacy is discussed, along with their relevance to the field of MOP receptor pharmacology, and in particular in relation to biased agonism at this important GPCR.
Collapse
Affiliation(s)
- E Kelly
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK.
| |
Collapse
|
36
|
Gabl M, Winther M, Skovbakke SL, Bylund J, Dahlgren C, Forsman H. A pepducin derived from the third intracellular loop of FPR2 is a partial agonist for direct activation of this receptor in neutrophils but a full agonist for cross-talk triggered reactivation of FPR2. PLoS One 2014; 9:e109516. [PMID: 25303226 PMCID: PMC4193777 DOI: 10.1371/journal.pone.0109516] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 09/02/2014] [Indexed: 11/18/2022] Open
Abstract
We recently described a novel receptor cross-talk mechanism in neutrophils, unique in that the signals generated by the PAF receptor (PAFR) and the ATP receptor (P2Y2R) transfer formyl peptide receptor 1 (FPR1) from a desensitized (non-signaling) state back to an actively signaling state (Forsman H et al., PLoS One, 8:e60169, 2013; Önnheim K, et al., Exp Cell Res, 323∶209, 2014). In addition to the G-protein coupled FPR1, neutrophils also express the closely related receptor FPR2. In this study we used an FPR2 specific pepducin, proposed to work as an allosteric modulator at the cytosolic signaling interface, to determine whether the cross-talk pathway is utilized also by FPR2. The pepducin used contains a fatty acid linked to a peptide sequence derived from the third intracellular loop of FPR2, and it activates as well as desensensitizes this receptor. We now show that neutrophils desensitized with the FPR2-specific pepducin display increased cellular responses to stimulation with PAF or ATP. The secondary PAF/ATP induced response was sensitive to FPR2-specific inhibitors, disclosing a receptor cross-talk mechanism underlying FPR2 reactivation. The pepducin induced an activity in naïve cells similar to that of a conventional FPR2 agonist, but with lower potency (partial efficacy), meaning that the pepducin is a partial agonist. The PAF- or ATP-induced reactivation was, however, much more pronounced when neutrophils had been desensitized to the pepducin as compared to cells desensitized to conventional agonists. The pepducin should thus in this respect be classified as a full agonist. In summary, we demonstrate that desensitized FPR2 can be transferred back to an actively signaling state by receptor cross-talk signals generated through PAFR and P2Y2R, and the difference in agonist potency with respect to pepducin-induced direct receptor activation and cross-talk reactivation of FPR2 puts the concept of functional selectivity in focus.
Collapse
Affiliation(s)
- Michael Gabl
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Malene Winther
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Sarah Line Skovbakke
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johan Bylund
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Claes Dahlgren
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Huamei Forsman
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
- * E-mail:
| |
Collapse
|
37
|
Sivertsen B, Holliday N, Madsen AN, Holst B. Functionally biased signalling properties of 7TM receptors - opportunities for drug development for the ghrelin receptor. Br J Pharmacol 2014; 170:1349-62. [PMID: 24032557 DOI: 10.1111/bph.12361] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 06/17/2013] [Accepted: 08/06/2013] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED The ghrelin receptor is a 7 transmembrane (7TM) receptor involved in a variety of physiological functions including growth hormone secretion, increased food intake and fat accumulation as well as modulation of reward and cognitive functions. Because of its important role in metabolism and energy expenditure, the ghrelin receptor has become an important therapeutic target for drug design and the development of anti-obesity compounds. However, none of the compounds developed so far have been approved for commercial use. Interestingly, the ghrelin receptor is able to signal through several different signalling pathways including Gαq , Gαi/o , Gα12/13 and arrestin recruitment. These multiple signalling pathways allow for functionally biased signalling, where one signalling pathway may be favoured over another either by selective ligands or through mutations in the receptor. In the present review, we have described how ligands and mutations in the 7TM receptor may bias the receptors to favour either one G-protein over another or to promote G-protein independent signalling pathways rather than G-protein-dependent pathways. For the ghrelin receptor, both agonist and inverse agonists have been demonstrated to signal more strongly through the Gαq -coupled pathway than the Gα12/13 -coupled pathway. Similarly a ligand that promotes Gαq coupling over Gαi coupling has been described and it has been suggested that several different active conformations of the receptor may exist dependent on the properties of the agonist. Importantly, ligands with such biased signalling properties may allow the development of drugs that selectively modulate only the therapeutically relevant physiological functions, thereby decreasing the risk of side effects. LINKED ARTICLES This article is part of a themed section on Neuropeptides. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.170.issue-7.
Collapse
Affiliation(s)
- B Sivertsen
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, the Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
38
|
Thompson GL, Kelly E, Christopoulos A, Canals M. Novel GPCR paradigms at the μ-opioid receptor. Br J Pharmacol 2014; 172:287-96. [PMID: 24460711 DOI: 10.1111/bph.12600] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/12/2014] [Accepted: 01/19/2014] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED Opioids, such as morphine, are the most clinically useful class of analgesic drugs for the treatment of acute and chronic pain. However, the use of opioids is greatly limited by the development of severe adverse side effects. Consequently, drug discovery efforts have been directed towards improving the therapeutic profile of opioid-based treatments. Opioid receptors are members of the family of GPCRs. As such, the recent GPCR paradigms of biased agonism and allosterism may provide novel avenues for more effective analgesics. Biased agonism (or functional selectivity) has been described for all the opioid receptor family members. Furthermore, the first allosteric modulators of opioid receptors have very recently been described. However, identification and quantification of biased agonism in a manner that is informative to medicinal chemists and drug discovery programmes still remains a challenge. In this review, we examine the progress, to date, towards identification and quantification of biased agonism and allosterism at the μ-opioid receptor in the context of its implications for the discovery of better and safer analgesics. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- G L Thompson
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| | | | | | | |
Collapse
|
39
|
Muscarinic acetylcholine receptors: novel opportunities for drug development. Nat Rev Drug Discov 2014; 13:549-60. [PMID: 24903776 DOI: 10.1038/nrd4295] [Citation(s) in RCA: 318] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The muscarinic acetylcholine receptors are a subfamily of G protein-coupled receptors that regulate numerous fundamental functions of the central and peripheral nervous system. The past few years have witnessed unprecedented new insights into muscarinic receptor physiology, pharmacology and structure. These advances include the first structural views of muscarinic receptors in both inactive and active conformations, as well as a better understanding of the molecular underpinnings of muscarinic receptor regulation by allosteric modulators. These recent findings should facilitate the development of new muscarinic receptor subtype-selective ligands that could prove to be useful for the treatment of many severe pathophysiological conditions.
Collapse
|
40
|
Szabo M, Klein Herenbrink C, Christopoulos A, Lane JR, Capuano B. Structure-activity relationships of privileged structures lead to the discovery of novel biased ligands at the dopamine D₂ receptor. J Med Chem 2014; 57:4924-39. [PMID: 24827597 DOI: 10.1021/jm500457x] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Biased agonism at GPCRs highlights the potential for the discovery and design of pathway-selective ligands and may confer therapeutic advantages to ligands targeting the dopamine D2 receptor (D2R). We investigated the determinants of efficacy, affinity, and bias for three privileged structures for the D2R, exploring changes to linker length and incorporation of a heterocyclic unit. Profiling the compounds in two signaling assays (cAMP and pERK1/2) allowed us to identify and quantify determinants of biased agonism at the D2R. Substitution on the phenylpiperazine privileged structures (2-methoxy vs 2,3-dichloro) influenced bias when the thienopyridine heterocycle was absent. Upon inclusion of the thienopyridine unit, the substitution pattern (4,6-dimethyl vs 5-chloro-6-methoxy-4-methyl) had a significant effect on bias that overruled the effect of the phenylpiperazine substitution pattern. This latter observation could be reconciled with an extended binding mode for these compounds, whereby the interaction of the heterocycle with a secondary binding pocket may engender bias.
Collapse
Affiliation(s)
- Monika Szabo
- Medicinal Chemistry and ‡Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville 3052, Victoria, Australia
| | | | | | | | | |
Collapse
|
41
|
Shonberg J, Lopez L, Scammells PJ, Christopoulos A, Capuano B, Lane JR. Biased Agonism at G Protein-Coupled Receptors: The Promise and the Challenges-A Medicinal Chemistry Perspective. Med Res Rev 2014; 34:1286-330. [DOI: 10.1002/med.21318] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jeremy Shonberg
- Medicinal Chemistry; Monash Institute of Pharmaceutical Sciences; Monash University (Parkville Campus); Parkville Victoria Australia
| | - Laura Lopez
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University (Parkville Campus); Parkville Victoria Australia
| | - Peter J. Scammells
- Medicinal Chemistry; Monash Institute of Pharmaceutical Sciences; Monash University (Parkville Campus); Parkville Victoria Australia
| | - Arthur Christopoulos
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University (Parkville Campus); Parkville Victoria Australia
| | - Ben Capuano
- Medicinal Chemistry; Monash Institute of Pharmaceutical Sciences; Monash University (Parkville Campus); Parkville Victoria Australia
| | - J. Robert Lane
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University (Parkville Campus); Parkville Victoria Australia
| |
Collapse
|
42
|
Correll CC, McKittrick BA. Biased ligand modulation of seven transmembrane receptors (7TMRs): functional implications for drug discovery. J Med Chem 2014; 57:6887-96. [PMID: 24697360 DOI: 10.1021/jm401677g] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Seven transmembrane receptors (7TMRs), also known as G-protein-coupled receptors (GPCRs), have proven to be valuable targets for the development of therapeutics. The expansion of our understanding of 7TMR downstream signaling pathways beyond G-proteins has broadened our appreciation of the versatility of these cell surface receptors. In particular, the increased awareness of 7TMR engagement of β-arrestin signaling has opened up additional avenues for drug discovery. 7TMRs can adopt different conformations and in response to various ligands can lead to a bias in downstream signaling mechanisms when comparing the overall efficacy between G-protein and β-arrestin dependent pathways. In 2012, we organized a session at the Spring National Meeting of the American Chemical Society on biased signaling in 7TMRs.1-4 Building on that experience, we provide in this Miniperspective some examples that exemplify developments in the area of biased 7TMR signaling and highlight some cautionary notes as well as some of the exciting opportunities for drug discovery.
Collapse
Affiliation(s)
- Craig C Correll
- Department of Immunology, Merck Research Laboratories , BMB 10-108, 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | | |
Collapse
|
43
|
Schann S, Bouvier M, Neuville P. Technology combination to address GPCR allosteric modulator drug-discovery pitfalls. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 10:e261-7. [PMID: 24050277 DOI: 10.1016/j.ddtec.2012.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Allosteric modulators (AMs) represent a novel paradigm to therapeutically target G-protein-coupled receptors (GPCRs). However, their identification and characterization using standard functional assays remain elusive due to the ‘context-dependent phenomena’. Novel technological approaches such as combining a Fluorescence Resonance Energy Transfer (FRET)-based library filtering with a Bioluminescence Resonance Energy Transfer (BRET)-based multiparametric compound profiling can circumvent the limitations of current GPCR screening processes and simplify the discovery of biased AMs.
Collapse
|
44
|
Kandola MK, Sykes L, Lee YS, Johnson MR, Hanyaloglu AC, Bennett PR. EP2 receptor activates dual G protein signaling pathways that mediate contrasting proinflammatory and relaxatory responses in term pregnant human myometrium. Endocrinology 2014; 155:605-17. [PMID: 24265450 DOI: 10.1210/en.2013-1761] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Prostaglandin (PG) E2 (PGE(2)) plays a central role in the regulation of smooth muscle contractions. Classically, PGE(2) stimulates contractions via EP1 and EP3 receptors, whereas EP2 and EP4 maintain quiescence. Labor involves a change from myometrial quiescence to contractions with a shift from anti- to proinflammatory pathways. EP2, a Gαs-coupled receptor, is known to mediate its actions via cAMP signaling. However, we have recently shown that EP2 also activates the proinflammatory PG G/H synthase-2 (PGHS-2). Here, we identify the mechanism underlying the ability of EP2 to maintain uterine quiescence and activate a proinflammatory/prolabor response in term-pregnant human myometrium. Human myometrial biopsies for in vivo and in vitro studies were taken at cesarean section at term, before or after the onset of labor. Activation of EP2 increased intracellular levels of cAMP and reduced contractility. Contrastingly, EP2 stimulation increased levels of PGHS-2, membrane-associated PGE synthase-1, and PGE(2). This was entirely dependent on EP2-mediated activation of calcium signaling. Both calcium signaling and up-regulation of PGHS-2 were insensitive to the Gαi inhibitor pertussis toxin but inhibited by small interfering RNA knockdown of Gαq/11. There were no differences in EP2 mRNA or protein levels between upper or lower segment myometrium or between pre- and postlabor myometrium. However, in myocytes taken after the onset of labor, cAMP signaling was markedly attenuated, whereas activation of calcium and PGHS-2 was preserved. Overall, the dual coupling of EP2 to Gαs-cAMP and Gαq/11-calcium pathways underlies its ability to mediate contrasting functions in term pregnancy and the "switching" to a prolabor receptor.
Collapse
Affiliation(s)
- Mandeep K Kandola
- Parturition Research (M.K.K., L.S., Y.S.L., M.R.J., P.R.B.) and G-Protein Coupled Receptor (GPCR) Signaling Groups (A.C.H.), Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, London W12 0NN, United Kingdom
| | | | | | | | | | | |
Collapse
|
45
|
Jean-Alphonse F, Bowersox S, Chen S, Beard G, Puthenveedu MA, Hanyaloglu AC. Spatially restricted G protein-coupled receptor activity via divergent endocytic compartments. J Biol Chem 2013; 289:3960-77. [PMID: 24375413 PMCID: PMC3924264 DOI: 10.1074/jbc.m113.526350] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Postendocytic sorting of G protein-coupled receptors (GPCRs) is driven by their interactions between highly diverse receptor sequence motifs with their interacting proteins, such as postsynaptic density protein (PSD95), Drosophila disc large tumor suppressor (Dlg1), zonula occludens-1 protein (zo-1) (PDZ) domain proteins. However, whether these diverse interactions provide an underlying functional specificity, in addition to driving sorting, is unknown. Here we identify GPCRs that recycle via distinct PDZ ligand/PDZ protein pairs that exploit their recycling machinery primarily for targeted endosomal localization and signaling specificity. The luteinizing hormone receptor (LHR) and β2-adrenergic receptor (B2AR), two GPCRs sorted to the regulated recycling pathway, underwent divergent trafficking to distinct endosomal compartments. Unlike B2AR, which traffics to early endosomes (EE), LHR internalizes to distinct pre-early endosomes (pre-EEs) for its recycling. Pre-EE localization required interactions of the LHR C-terminal tail with the PDZ protein GAIP-interacting protein C terminus, inhibiting its traffic to EEs. Rerouting the LHR to EEs, or EE-localized GPCRs to pre-EEs, spatially reprograms MAPK signaling. Furthermore, LHR-mediated activation of MAPK signaling requires internalization and is maintained upon loss of the EE compartment. We propose that combinatorial specificity between GPCR sorting sequences and interacting proteins dictates an unprecedented spatiotemporal control in GPCR signal activity.
Collapse
Affiliation(s)
- Frederic Jean-Alphonse
- From the Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, London W12 0NN, United Kingdom and
| | | | | | | | | | | |
Collapse
|
46
|
Pepducin targeting the C-X-C chemokine receptor type 4 acts as a biased agonist favoring activation of the inhibitory G protein. Proc Natl Acad Sci U S A 2013; 110:E5088-97. [PMID: 24309376 DOI: 10.1073/pnas.1312515110] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Short lipidated peptide sequences derived from various intracellular loop regions of G protein-coupled receptors (GPCRs) are named pepducins and act as allosteric modulators of a number of GPCRs. Recently, a pepducin selectively targeting the C-X-C chemokine receptor type 4 (CXCR4) was found to be an allosteric agonist, active in both cell-based assays and in vivo. However, the precise mechanism of action of this class of ligands remains poorly understood. In particular, given the diversity of signaling effectors that can be engaged by a given receptor, it is not clear whether pepducins can show biased signaling leading to functional selectivity. To explore the ligand-biased potential of pepducins, we assessed the effect of the CXCR4 selective pepducin, ATI-2341, on the ability of the receptor to engage the inhibitory G proteins (Gi1, Gi2 and Gi3), G13, and β-arrestins. Using bioluminescence resonance energy transfer-based biosensors, we found that, in contrast to the natural CXCR4 ligand, stromal cell-derived factor-1α, which promotes the engagement of the three Gi subtypes, G13 and the two β-arrestins, ATI-2341 leads to the engagement of the Gi subtypes but not G13 or the β-arrestins. Calculation of the transduction ratio for each pathway revealed a strong negative bias of ATI-2341 toward G13 and β-arrestins, revealing functional selectivity for the Gi pathways. The negative bias toward β-arrestins results from the reduced ability of the pepducin to promote GPCR kinase-mediated phosphorylation of the receptor. In addition to revealing ligand-biased signaling of pepducins, these findings shed some light on the mechanism of action of a unique class of allosteric regulators.
Collapse
|
47
|
Shonberg J, Herenbrink CK, López L, Christopoulos A, Scammells PJ, Capuano B, Lane JR. A structure-activity analysis of biased agonism at the dopamine D2 receptor. J Med Chem 2013; 56:9199-221. [PMID: 24138311 DOI: 10.1021/jm401318w] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Biased agonism offers an opportunity for the medicinal chemist to discover pathway-selective ligands for GPCRs. A number of studies have suggested that biased agonism at the dopamine D2 receptor (D2R) may be advantageous for the treatment of neuropsychiatric disorders, including schizophrenia. As such, it is of great importance to gain insight into the SAR of biased agonism at this receptor. We have generated SAR based on a novel D2R partial agonist, tert-butyl (trans-4-(2-(3,4-dihydroisoquinolin-2(1H)-yl)ethyl)cyclohexyl)carbamate (4). This ligand shares structural similarity to cariprazine (2), a drug awaiting FDA approval for the treatment of schizophrenia, yet displays a distinct bias toward two different signaling end points. We synthesized a number of derivatives of 4 with subtle structural modifications, including incorporation of cariprazine fragments. By combining pharmacological profiling with analytical methodology to identify and to quantify bias, we have demonstrated that efficacy and biased agonism can be finely tuned by minor structural modifications to the head group containing the tertiary amine, a tail group that extends away from this moiety, and the orientation and length of a spacer region between these two moieties.
Collapse
Affiliation(s)
- Jeremy Shonberg
- Medicinal Chemistry, ‡Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus) , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | | | | | | | | | | | | |
Collapse
|
48
|
Ahn KH, Scott CE, Abrol R, Goddard WA, Kendall DA. Computationally-predicted CB1 cannabinoid receptor mutants show distinct patterns of salt-bridges that correlate with their level of constitutive activity reflected in G protein coupling levels, thermal stability, and ligand binding. Proteins 2013; 81:1304-17. [PMID: 23408552 DOI: 10.1002/prot.24264] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 12/21/2013] [Accepted: 01/21/2013] [Indexed: 11/09/2022]
Abstract
The cannabinoid receptor 1 (CB1), a member of the class A G-protein-coupled receptor (GPCR) family, possesses an observable level of constitutive activity. Its activation mechanism, however, has yet to be elucidated. Previously we discovered dramatic changes in CB1 activity due to single mutations; T3.46A, which made the receptor inactive, and T3.46I and L3.43A, which made it essentially fully constitutively active. Our subsequent prediction of the structures of these mutant receptors indicated that these changes in activity are explained in terms of the pattern of salt-bridges in the receptor region involving transmembrane domains 2, 3, 5, and 6. Here we identified key salt-bridges, R2.37 + D6.30 and D2.63 + K3.28, critical for CB1 inactive and active states, respectively, and generated new mutant receptors that we predicted would change CB1 activity by either precluding or promoting these interactions. We find that breaking the R2.37 + D6.30 salt-bridge resulted in substantial increase in G-protein coupling activity and reduced thermal stability relative to the wild-type reflecting the changes in constitutive activity from inactive to active. In contrast, breaking the D2.63 + K3.28 salt-bridge produced the opposite profile suggesting this interaction is critical for the receptor activation. Thus, we demonstrate an excellent correlation with the predicted pattern of key salt-bridges and experimental levels of activity and conformational flexibility. These results are also consistent with the extended ternary complex model with respect to shifts in agonist and inverse agonist affinity and provide a powerful framework for understanding the molecular basis for the multiple stages of CB1 activation and that of other GPCRs in general.
Collapse
Affiliation(s)
- Kwang H Ahn
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269-3092, USA
| | | | | | | | | |
Collapse
|
49
|
Abstract
INTRODUCTION There is an unmet need for a new class of direct bronchodilators for the treatment of asthma and chronic obstructive lung disease. Unexpectedly, bitter taste receptors (TAS2Rs) have been localized on airway smooth muscle and when activated cause marked smooth muscle relaxation through a mechanism that is distinct from β2-adrenegic receptors. Thus TAS2R agonists have emerged as a novel class of bronchodilator. AREAS COVERED A synopsis of the TAS2R family and its biology for bitter taste perception on the tongue is provided, followed by a review of the identification and molecular and physiological characterization of TAS2R subtypes on human and mouse airway smooth muscle. The proposed molecular mechanisms leading to the relaxation response are provided, along with gaps in our understanding at certain points in the signaling cascade. Unresolved issues that may need to be considered for drug development are discussed. EXPERT OPINION TAS2R agonists show promise as a new class of highly efficacious bronchodilators for treatment of obstructive lung disease. With tens of thousands of known natural and synthetic bitter compounds, there is substantial diversity within the known agonists, and, a ready source of agents for screening and further development of an inhaled TAS2R agonist for therapeutic purposes.
Collapse
Affiliation(s)
- Stephen B Liggett
- University of South Florida Morsani College of Medicine, Departments of Internal Medicine and Molecular Pharmacology and Physiology , 12901 Bruce B. Downs Blvd, MDC02 Tampa, FL 33612 , USA.
| |
Collapse
|
50
|
Abstract
The neoclerodane diterpene salvinorin A is the major active component of the hallucinogenic mint plant Salvia divinorum Epling and Játiva (Lamiaceae). Since the finding that salvinorin A exerts its potent psychotropic actions through the activation of opioid receptors, the site of action of morphine and related analogues, there has been much interest in elucidating the underlying mechanisms behind its effects. These effects are particularly remarkable because (1) salvinorin A is the first reported non-nitrogenous opioid receptor agonist and (2) its effects are not mediated through the previously investigated targets of psychotomimetics. This Perspective outlines our research program, illustrating a new direction to the development of tools to further elucidate the biological mechanisms of drug tolerance and dependence. The information gained from these efforts is expected to facilitate the design of novel agents to treat pain, drug abuse, and other central nervous system disorders.
Collapse
Affiliation(s)
- Thomas E Prisinzano
- Department of Medicinal Chemistry, University of Kansas , 1251 Wescoe Hall Drive, 4070 Malott Hall, Lawrence, Kansas 66045-7572, United States.
| |
Collapse
|