1
|
Crecelius JM, Manz AR, Benzow S, Marchese A. Receptor Determinants for β-Arrestin Functional Specificity at C-X-C Chemokine Receptor 5. Mol Pharmacol 2024; 106:287-297. [PMID: 39472027 PMCID: PMC11585254 DOI: 10.1124/molpharm.124.000942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/07/2024] [Indexed: 11/20/2024] Open
Abstract
β-arrestins are multifaceted adaptor proteins that mediate G protein-coupled receptor (GPCR) desensitization, internalization, and signaling. It is emerging that receptor-specific determinants specify these divergent functions at GPCRs, yet this remains poorly understood. Here, we set out to identify the receptor determinants responsible for β-arrestin-mediated regulation of the chemokine receptor C-X-C motif chemokine receptor 5 (CXCR5). Using bioluminescence resonance energy transfer, we show that β-arrestin1 and β-arrestin2 are dose-dependently recruited to CXCR5 by its cognate ligand C-X-C motif chemokine ligand 13 (CXCL13). The carboxy-terminal tail of CXCR5 contains several serine/threonine residues that can be divided into three discrete phospho-site clusters based on their position relative to transmembrane domain 7. Mutagenesis experiments revealed that the distal and medial phospho-site clusters, but not the proximal, are required for agonist-stimulated β-arrestin1 or β-arrestin2 recruitment to CXCR5. Consistent with this, we provide evidence that the distal and medial, but not proximal, phospho-site clusters are required for receptor desensitization. Surprisingly, the individual phospho-site clusters are not required for agonist-stimulated internalization of CXCR5. Further, we show that CXCL13-stimulated CXCR5 internalization and ERK1/2 phosphorylation, but not desensitization, remain intact in human embryonic kidney 293 cells lacking β-arrestin1 and β-arrestin2. Our study provides evidence that β-arrestins are recruited to CXCR5 and are required for desensitization but are dispensable for internalization or signaling, suggesting that discrete receptor determinants specify the divergent functions of β-arrestins. SIGNIFICANCE STATEMENT: C-X-C motif ligand 13 (CXCL13) and C-X-C motif chemokine receptor 5 (CXCR5) are important in the immune system and are linked to diseases, yet regulation of CXCR5 signaling remains poorly understood. We provide evidence that a phospho-site cluster located at the extreme distal carboxyl-terminal tail of the receptor is responsible for β-arrestin recruitment and receptor desensitization. β-arrestins are not required for CXCL13-stimulated internalization or signaling, indicating that β-arrestins perform only one of their functions at CXCR5 and that discrete receptor determinants specify the divergent functions of β-arrestins.
Collapse
Affiliation(s)
- Joseph M Crecelius
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Aaren R Manz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sara Benzow
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
2
|
Zheng C, Javitch JA, Lambert NA, Donthamsetti P, Gurevich VV. In-Cell Arrestin-Receptor Interaction Assays. Curr Protoc 2023; 3:e890. [PMID: 37787634 PMCID: PMC10566372 DOI: 10.1002/cpz1.890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
G protein-coupled receptors (GPCRs) represent ∼30% of current drug targets. Ligand binding to these receptors activates G proteins and arrestins, which function in different signaling pathways. Given that functionally selective or biased ligands preferentially activate one of these two groups of pathways, they may be superior medications for certain disease states. The identification of such ligands requires robust drug screening assays for both G protein and arrestin activity. This unit describes protocols for assays that monitor reversible arrestin recruitment to GPCRs in living cells using either bioluminescence resonance energy transfer (BRET) or nanoluciferase complementation (NanoLuc). Two types of assays can be used: one configuration directly measures arrestin recruitment to a GPCR fused to a protein tag at its intracellular C-terminus, whereas the other configuration detects arrestin translocation to the plasma membrane in response to activation of an unmodified GPCR. Together, these assays are powerful tools for studying dynamic interactions between GPCRs and arrestins. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Receptor-arrestin BRET assay to measure ligand-induced recruitment of arrestin to receptors Basic Protocol 2: Receptor-arrestin NANOBIT assay to measure ligand-induced recruitment of arrestin to receptors Alternative Protocol 1: BRET assay to measure ligand-induced recruitment of arrestin to the plasma membrane Alternative Protocol 2: NANOBIT assay to measure ligand-induced recruitment of arrestin to the plasma membrane Support Protocol 1: Optimization of polyethylenimine (PEI) concentration for transfection.
Collapse
Affiliation(s)
- Chen Zheng
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Jonathan A. Javitch
- Departments of Psychiatry and Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York
| | - Nevin A. Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | | | | |
Collapse
|
3
|
Fouillen A, Bous J, Granier S, Mouillac B, Sounier R. Bringing GPCR Structural Biology to Medical Applications: Insights from Both V2 Vasopressin and Mu-Opioid Receptors. MEMBRANES 2023; 13:606. [PMID: 37367810 PMCID: PMC10303988 DOI: 10.3390/membranes13060606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023]
Abstract
G-protein coupled receptors (GPCRs) are versatile signaling proteins that regulate key physiological processes in response to a wide variety of extracellular stimuli. The last decade has seen a revolution in the structural biology of clinically important GPCRs. Indeed, the improvement in molecular and biochemical methods to study GPCRs and their transducer complexes, together with advances in cryo-electron microscopy, NMR development, and progress in molecular dynamic simulations, have led to a better understanding of their regulation by ligands of different efficacy and bias. This has also renewed a great interest in GPCR drug discovery, such as finding biased ligands that can either promote or not promote specific regulations. In this review, we focus on two therapeutically relevant GPCR targets, the V2 vasopressin receptor (V2R) and the mu-opioid receptor (µOR), to shed light on the recent structural biology studies and show the impact of this integrative approach on the determination of new potential clinical effective compounds.
Collapse
Affiliation(s)
- Aurélien Fouillen
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34000 Montpellier, France; (A.F.); (S.G.); (B.M.)
- Centre de Biochimie Structurale (CBS), Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France
| | - Julien Bous
- Section of Receptor Biology & Signaling, Department of Physiology & Pharmacology, Karolinska Institutet, 17165 Stockholm, Sweden;
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34000 Montpellier, France; (A.F.); (S.G.); (B.M.)
| | - Bernard Mouillac
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34000 Montpellier, France; (A.F.); (S.G.); (B.M.)
| | - Remy Sounier
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34000 Montpellier, France; (A.F.); (S.G.); (B.M.)
| |
Collapse
|
4
|
Ehrlich AT, Couvineau P, Schamiloglu S, Wojcik S, Da Fonte D, Mezni A, von Zastrow M, Bender KJ, Bouvier M, Kieffer BL. Visualization of real-time receptor endocytosis in dopamine neurons enabled by NTSR1-Venus knock-in mice. Front Cell Neurosci 2022; 16:1076599. [DOI: 10.3389/fncel.2022.1076599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/11/2022] [Indexed: 11/30/2022] Open
Abstract
Dopamine (DA) neurons are primarily concentrated in substantia nigra (SN) and ventral tegmental area (VTA). A subset of these neurons expresses the neurotensin receptor NTSR1 and its putative ligand neurotensin (Nts). NTSR1, a G protein-coupled receptor (GPCR), which classically activates Gαq/calcium signaling, is a potential route for modulating DA activity. Drug development efforts have been hampered by the receptor’s complex pharmacology and a lack of understanding about its endogenous location and signaling responses. Therefore, we have generated NTSR1-Venus knock-in (KI) mice to study NTSR1 receptors in their physiological context. In primary hippocampal neurons, we show that these animals express functional receptors that respond to agonists by increasing intracellular calcium release and trafficking to endosomes. Moreover, systemic agonist administration attenuates locomotion in KIs as it does in control animals. Mapping receptor protein expression at regional and cellular levels, located NTSR1-Venus on the soma and dendrites of dopaminergic SN/VTA neurons. Direct monitoring of receptor endocytosis, as a proxy for activation, enabled profiling of NTSR1 agonists in neurons, as well as acute SN/VTA containing brain slices. Taken together, NTSR1-Venus animals express traceable receptors that will improve understanding of NTSR1 and DA activities and more broadly how GPCRs act in vivo.
Collapse
|
5
|
Yu Z, Cary BP, Kim TW, Nguyen KD, Gardella TJ, Gellman SH. Kinetic and Thermodynamic Insights into Agonist Interactions with the Parathyroid Hormone Receptor-1 from a New NanoBRET Assay. ACS Chem Biol 2022; 17:3148-3158. [PMID: 36282520 PMCID: PMC9747329 DOI: 10.1021/acschembio.2c00595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Polypeptides that activate the parathyroid hormone receptor-1 (PTHR1) are important in human physiology and medicine. Most previous studies of peptide binding to this receptor have involved the displacement of a radiolabeled ligand. We report a new assay format based on bioluminescence resonance energy transfer (BRET). Fusion of a NanoLuc luciferase (nLuc) unit to the N-terminus of the PTHR1 allows the direct detection of binding by an agonist peptide bearing a tetramethylrhodamine (TMR) unit. Affinity measurements from the BRET assay align well with results previously obtained via radioligand displacement. The BRET assay offers substantial operational benefits relative to affinity measurements involving radioactive compounds. The convenience of the new assay allowed us to explore several questions raised by earlier reports. For example, we show that although the first two residues of PTH(1-34) (the drug teriparatide) are critical for PTHR1 activation, these two residues contribute little or nothing to affinity. Comparisons among the well-studied agonists PTH(1-34), PTHrP(1-34), and "long-acting PTH" (LA-PTH) reveal that the high affinity of LA-PTH arises largely from a diminished rate constant for dissociation relative to the other two. A D-peptide recently reported to be comparable to PTH(1-34) as an agonist of the PTHR1 was found not to bind detectably to the receptor and to be a very weak agonist.
Collapse
Affiliation(s)
- Zhen Yu
- Department of Chemistry, University of Wisconsin - Madison, Madison, WI 53706 USA
| | - Brian P. Cary
- Department of Chemistry, University of Wisconsin - Madison, Madison, WI 53706 USA
| | - Tae Wook Kim
- Department of Chemistry, University of Wisconsin - Madison, Madison, WI 53706 USA
| | - Kevin D. Nguyen
- Department of Chemistry, University of Wisconsin - Madison, Madison, WI 53706 USA
| | - Thomas J. Gardella
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA
| | - Samuel H. Gellman
- Department of Chemistry, University of Wisconsin - Madison, Madison, WI 53706 USA
| |
Collapse
|
6
|
Guo S, Zhao T, Yun Y, Xie X. Recent Progress in Assays for GPCR Drug Discovery. Am J Physiol Cell Physiol 2022; 323:C583-C594. [PMID: 35816640 DOI: 10.1152/ajpcell.00464.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
G-protein coupled receptors (GPCRs), also known as 7 transmembrane receptors, are the largest family of cell surface receptors in eukaryotes. There are ~800 GPCRs in human, regulating diverse physiological processes. GPCRs are the most intensively studied drug targets. Drugs that target GPCRs account for about a quarter of the global market share of therapeutic drugs. Therefore, to develop physiologically relevant and robust assays to search new GPCR ligands or modulators remain the major focus of drug discovery research worldwide. Early functional GPCR assays are mainly depend on the measurement of G protein-mediated second messenger generation. Recent development in GPCR biology indicate the signaling of these receptors is much more complex than the oversimplified classical view. GPCRs have been found to activate multiple G proteins simultaneously and induce b-arrestin-mediated signaling. GPCRs have also been found to interacte with other cytosolic scaffolding proteins and form dimer or heteromer with GPCRs or other transmembrane proteins. Here we mainly discuss technologies focused on detecting protein-protein interactions, such as FRET/BRET, NanoBiT, Tango, etc, and their applications in measuring GPCRs interacting with various signaling partners. In the final part, we also discuss the species differences in GPCRs when using animal models to study the in vivofunctions of GPCR ligands, and possible ways to solve this problem with modern genetic tools.
Collapse
Affiliation(s)
- Shimeng Guo
- grid.419093.6Shanghai Institute of Materia Medica, Shanghai, China
| | - Tingting Zhao
- grid.419093.6Shanghai Institute of Materia Medica, Shanghai, China
| | - Ying Yun
- grid.419093.6Shanghai Institute of Materia Medica, Shanghai, China
| | - Xin Xie
- grid.419093.6Shanghai Institute of Materia Medica, Shanghai, China
| |
Collapse
|
7
|
A Real-Time, Plate-Based BRET Assay for Detection of cGMP in Primary Cells. Int J Mol Sci 2022; 23:ijms23031908. [PMID: 35163827 PMCID: PMC8837005 DOI: 10.3390/ijms23031908] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/30/2022] [Accepted: 02/04/2022] [Indexed: 11/30/2022] Open
Abstract
Cyclic guanosine monophosphate (cGMP) is a second messenger involved in the regulation of numerous physiological processes. The modulation of cGMP is important in many diseases, but reliably assaying cGMP in live cells in a plate-based format with temporal resolution is challenging. The Förster/fluorescence resonance energy transfer (FRET)-based biosensor cGES-DE5 has a high temporal resolution and high selectivity for cGMP over cAMP, so we converted it to use bioluminescence resonance energy transfer (BRET), which is more compatible with plate-based assays. This BRET variant, called CYGYEL (cyclic GMP sensor using YFP-PDE5-Rluc8), was cloned into a lentiviral vector for use across different mammalian cell types. CYGYEL was characterised in HEK293T cells using the nitric oxide donor diethylamine NONOate (DEA), where it was shown to be dynamic, reversible, and able to detect cGMP with or without the use of phosphodiesterase inhibitors. In human primary vascular endothelial and smooth muscle cells, CYGYEL successfully detected cGMP mediated through either soluble or particulate guanylate cyclase using DEA or C-type natriuretic peptide, respectively. Notably, CYGYEL detected differences in kinetics and strength of signal both between ligands and between cell types. CYGYEL remained selective for cGMP over cAMP, but this selectivity was reduced compared to cGES-DE5. CYGYEL streamlines the process of cGMP detection in plate-based assays and can be used to detect cGMP activity across a range of cell types.
Collapse
|
8
|
Ehrlich AT, Semache M, Couvineau P, Wojcik S, Kobayashi H, Thelen M, Gross F, Hogue M, Le Gouill C, Darcq E, Bouvier M, Kieffer BL. Ackr3-Venus knock-in mouse lights up brain vasculature. Mol Brain 2021; 14:151. [PMID: 34583741 PMCID: PMC8477500 DOI: 10.1186/s13041-021-00862-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/17/2021] [Indexed: 01/09/2023] Open
Abstract
The atypical chemokine receptor 3, ACKR3, is a G protein-coupled receptor, which does not couple to G proteins but recruits βarrestins. At present, ACKR3 is considered a target for cancer and cardiovascular disorders, but less is known about the potential of ACKR3 as a target for brain disease. Further, mouse lines have been created to identify cells expressing the receptor, but there is no tool to visualize and study the receptor itself under physiological conditions. Here, we engineered a knock-in (KI) mouse expressing a functional ACKR3-Venus fusion protein to directly detect the receptor, particularly in the adult brain. In HEK-293 cells, native and fused receptors showed similar membrane expression, ligand induced trafficking and signaling profiles, indicating that the Venus fusion does not alter receptor signaling. We also found that ACKR3-Venus enables direct real-time monitoring of receptor trafficking using resonance energy transfer. In ACKR3-Venus knock-in mice, we found normal ACKR3 mRNA levels in the brain, suggesting intact gene transcription. We fully mapped receptor expression across 14 peripheral organs and 112 brain areas and found that ACKR3 is primarily localized to the vasculature in these tissues. In the periphery, receptor distribution aligns with previous reports. In the brain there is notable ACKR3 expression in endothelial vascular cells, hippocampal GABAergic interneurons and neuroblast neighboring cells. In conclusion, we have generated Ackr3-Venus knock-in mice with a traceable ACKR3 receptor, which will be a useful tool to the research community for interrogations about ACKR3 biology and related diseases.
Collapse
Affiliation(s)
- Aliza T Ehrlich
- Douglas Research Center, McGill University, Montréal, Canada.
- University of California, San Francisco, USA.
| | - Meriem Semache
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
- Domain Therapeutics North America, Montréal, Québec, H4S 1Z9, Canada
| | - Pierre Couvineau
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Stefan Wojcik
- Douglas Research Center, McGill University, Montréal, Canada
- University of Surrey, Guildford, UK
- Oxford Brookes University, Oxford, UK
| | - Hiroyuki Kobayashi
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Florence Gross
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
- Domain Therapeutics North America, Montréal, Québec, H4S 1Z9, Canada
| | - Mireille Hogue
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Christian Le Gouill
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Emmanuel Darcq
- Douglas Research Center, McGill University, Montréal, Canada
- INSERM U1114, University of Strasbourg, Strasbourg, France
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada.
| | - Brigitte L Kieffer
- Douglas Research Center, McGill University, Montréal, Canada.
- INSERM U1114, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
9
|
Schihada H, Shekhani R, Schulte G. Quantitative assessment of constitutive G protein-coupled receptor activity with BRET-based G protein biosensors. Sci Signal 2021; 14:eabf1653. [PMID: 34516756 DOI: 10.1126/scisignal.abf1653] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hannes Schihada
- Section for Receptor Biology and Signaling, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17165 Stockholm, Sweden
| | - Rawan Shekhani
- Section for Receptor Biology and Signaling, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17165 Stockholm, Sweden
| | - Gunnar Schulte
- Section for Receptor Biology and Signaling, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17165 Stockholm, Sweden
| |
Collapse
|
10
|
Lotz-Havla AS, Woidy M, Guder P, Friedel CC, Klingbeil JM, Bulau AM, Schultze A, Dahmen I, Noll-Puchta H, Kemp S, Erdmann R, Zimmer R, Muntau AC, Gersting SW. iBRET Screen of the ABCD1 Peroxisomal Network and Mutation-Induced Network Perturbations. J Proteome Res 2021; 20:4366-4380. [PMID: 34383492 DOI: 10.1021/acs.jproteome.1c00330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mapping the network of proteins provides a powerful means to investigate the function of disease genes and to unravel the molecular basis of phenotypes. We present an automated informatics-aided and bioluminescence resonance energy transfer-based approach (iBRET) enabling high-confidence detection of protein-protein interactions in living mammalian cells. A screen of the ABCD1 protein, which is affected in X-linked adrenoleukodystrophy (X-ALD), against an organelle library of peroxisomal proteins demonstrated applicability of iBRET for large-scale experiments. We identified novel protein-protein interactions for ABCD1 (with ALDH3A2, DAO, ECI2, FAR1, PEX10, PEX13, PEX5, PXMP2, and PIPOX), mapped its position within the peroxisomal protein-protein interaction network, and determined that pathogenic missense variants in ABCD1 alter the interaction with selected binding partners. These findings provide mechanistic insights into pathophysiology of X-ALD and may foster the identification of new disease modifiers.
Collapse
Affiliation(s)
- Amelie S Lotz-Havla
- Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| | - Mathias Woidy
- University Children's Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Philipp Guder
- University Children's Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Caroline C Friedel
- Institute of Informatics, Ludwig-Maximilians-Universität München, 80538 Munich, Germany
| | - Julian M Klingbeil
- Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| | - Ana-Maria Bulau
- Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| | - Anja Schultze
- Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| | - Ilona Dahmen
- Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| | - Heidi Noll-Puchta
- Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| | - Stephan Kemp
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, 1105 WX Amsterdam, The Netherlands
| | - Ralf Erdmann
- Systems Biochemistry, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Ralf Zimmer
- Institute of Informatics, Ludwig-Maximilians-Universität München, 80538 Munich, Germany
| | - Ania C Muntau
- University Children's Hospital, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Søren W Gersting
- University Children's Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
11
|
Zhou Y, Meng J, Xu C, Liu J. Multiple GPCR Functional Assays Based on Resonance Energy Transfer Sensors. Front Cell Dev Biol 2021; 9:611443. [PMID: 34041234 PMCID: PMC8141573 DOI: 10.3389/fcell.2021.611443] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/05/2021] [Indexed: 12/19/2022] Open
Abstract
G protein-coupled receptors (GPCRs) represent one of the largest membrane protein families that participate in various physiological and pathological activities. Accumulating structural evidences have revealed how GPCR activation induces conformational changes to accommodate the downstream G protein or β-arrestin. Multiple GPCR functional assays have been developed based on Förster resonance energy transfer (FRET) and bioluminescence resonance energy transfer (BRET) sensors to monitor the conformational changes in GPCRs, GPCR/G proteins, or GPCR/β-arrestin, especially over the past two decades. Here, we will summarize how these sensors have been optimized to increase the sensitivity and compatibility for application in different GPCR classes using various labeling strategies, meanwhile provide multiple solutions in functional assays for high-throughput drug screening.
Collapse
Affiliation(s)
- Yiwei Zhou
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jiyong Meng
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Chanjuan Xu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jianfeng Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
12
|
Abrimian A, Kraft T, Pan YX. Endogenous Opioid Peptides and Alternatively Spliced Mu Opioid Receptor Seven Transmembrane Carboxyl-Terminal Variants. Int J Mol Sci 2021; 22:3779. [PMID: 33917474 PMCID: PMC8038826 DOI: 10.3390/ijms22073779] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 12/20/2022] Open
Abstract
There exist three main types of endogenous opioid peptides, enkephalins, dynorphins and β-endorphin, all of which are derived from their precursors. These endogenous opioid peptides act through opioid receptors, including mu opioid receptor (MOR), delta opioid receptor (DOR) and kappa opioid receptor (KOR), and play important roles not only in analgesia, but also many other biological processes such as reward, stress response, feeding and emotion. The MOR gene, OPRM1, undergoes extensive alternative pre-mRNA splicing, generating multiple splice variants or isoforms. One type of these splice variants, the full-length 7 transmembrane (TM) Carboxyl (C)-terminal variants, has the same receptor structures but contains different intracellular C-terminal tails. The pharmacological functions of several endogenous opioid peptides through the mouse, rat and human OPRM1 7TM C-terminal variants have been considerably investigated together with various mu opioid ligands. The current review focuses on the studies of these endogenous opioid peptides and summarizes the results from early pharmacological studies, including receptor binding affinity and G protein activation, and recent studies of β-arrestin2 recruitment and biased signaling, aiming to provide new insights into the mechanisms and functions of endogenous opioid peptides, which are mediated through the OPRM1 7TM C-terminal splice variants.
Collapse
Affiliation(s)
| | | | - Ying-Xian Pan
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (A.A.); (T.K.)
| |
Collapse
|
13
|
Slosky LM, Caron MG, Barak LS. Biased Allosteric Modulators: New Frontiers in GPCR Drug Discovery. Trends Pharmacol Sci 2021; 42:283-299. [PMID: 33581873 PMCID: PMC9797227 DOI: 10.1016/j.tips.2020.12.005] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/31/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest class of cell surface receptors in the genome and the most successful family of targets of FDA-approved drugs. New frontiers in GPCR drug discovery remain, however, as achieving receptor subtype selectivity and controlling off- and on-target side effects are not always possible with classic agonist and antagonist ligands. These challenges may be overcome by focusing development efforts on allosteric ligands that confer signaling bias. Biased allosteric modulators (BAMs) are an emerging class of GPCR ligands that engage less well-conserved regulatory motifs outside the orthosteric pocket and exert pathway-specific effects on receptor signaling. The unique ways that BAMs texturize receptor signaling present opportunities to fine-tune physiology and develop safer, more selective therapeutics. Here, we provide a conceptual framework for understanding the pharmacology of BAMs, explore their therapeutic potential, and discuss strategies for their discovery.
Collapse
Affiliation(s)
- Lauren M. Slosky
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Marc G. Caron
- Departments of Cell Biology, Neurobiology and Medicine, Duke University, Durham, NC 27710, USA,Correspondence: (L.S.B.); (M.G.C.)
| | - Lawrence S. Barak
- Department of Cell Biology, Duke University, Durham, NC 27710, USA,Correspondence: (L.S.B.); (M.G.C.)
| |
Collapse
|
14
|
Aydin Y, Coin I. Biochemical insights into structure and function of arrestins. FEBS J 2021; 288:2529-2549. [DOI: 10.1111/febs.15811] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/26/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Yasmin Aydin
- Institute of Biochemistry Faculty of Life Sciences University of Leipzig Germany
| | - Irene Coin
- Institute of Biochemistry Faculty of Life Sciences University of Leipzig Germany
| |
Collapse
|
15
|
Moo EV, van Senten JR, Bräuner-Osborne H, Møller TC. Arrestin-Dependent and -Independent Internalization of G Protein-Coupled Receptors: Methods, Mechanisms, and Implications on Cell Signaling. Mol Pharmacol 2021; 99:242-255. [PMID: 33472843 DOI: 10.1124/molpharm.120.000192] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/07/2021] [Indexed: 01/05/2023] Open
Abstract
Agonist-induced endocytosis is a key regulatory mechanism for controlling the responsiveness of the cell by changing the density of cell surface receptors. In addition to the role of endocytosis in signal termination, endocytosed G protein-coupled receptors (GPCRs) have been found to signal from intracellular compartments of the cell. Arrestins are generally believed to be the master regulators of GPCR endocytosis by binding to both phosphorylated receptors and adaptor protein 2 (AP-2) or clathrin, thus recruiting receptors to clathrin-coated pits to facilitate the internalization process. However, many other functions have been described for arrestins that do not relate to their role in terminating signaling. Additionally, there are now more than 30 examples of GPCRs that internalize independently of arrestins. Here we review the methods, pharmacological tools, and cellular backgrounds used to determine the role of arrestins in receptor internalization, highlighting their advantages and caveats. We also summarize key examples of arrestin-independent GPCR endocytosis in the literature and their suggested alternative endocytosis pathway (e.g., the caveolae-dependent and fast endophilin-mediated endocytosis pathways). Finally, we consider the possible function of arrestins recruited to GPCRs that are endocytosed independently of arrestins, including the catalytic arrestin activation paradigm. Technological improvements in recent years have advanced the field further, and, combined with the important implications of endocytosis on drug responses, this makes endocytosis an obvious parameter to include in molecular pharmacological characterization of ligand-GPCR interactions. SIGNIFICANCE STATEMENT: G protein-coupled receptor (GPCR) endocytosis is an important means to terminate receptor signaling, and arrestins play a central role in the widely accepted classical paradigm of GPCR endocytosis. In contrast to the canonical arrestin-mediated internalization, an increasing number of GPCRs are found to be endocytosed via alternate pathways, and the process appears more diverse than the previously defined "one pathway fits all."
Collapse
Affiliation(s)
- Ee Von Moo
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Jeffrey R van Senten
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Thor C Møller
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Narayan A, Hunkele A, Xu J, Bassoni DL, Pasternak GW, Pan YX. Mu Opioids Induce Biased Signaling at the Full-Length Seven Transmembrane C-Terminal Splice Variants of the mu Opioid Receptor Gene, Oprm1. Cell Mol Neurobiol 2020; 41:1059-1074. [PMID: 33033993 DOI: 10.1007/s10571-020-00973-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/23/2020] [Indexed: 01/06/2023]
Abstract
The biased signaling has been extensively studied in the original mu opioid receptor (MOR-1), particularly through G protein and β-arrestin2 signaling pathways. The concept that the G protein pathway is often linked to the therapeutic effect of the drug, while the β-arrestin pathway is associated to the side effects has been proposed to develop biased analgesic compounds with limited side-effects associated with traditional opiates. The mu opioid receptor gene, OPRM1, undergoes extensive alternative pre-mRNA splicing, generating multiple splice variants or isoforms that are conserved from rodent to human. One type of the Oprm1 splice variants are the full-length 7 transmembrane (7TM) C-terminal splice variants, which have identical receptor structures including entire binding pocket, but contain a different intracellular C-terminal tail resulted from 3' alternative splicing. Increasing evidence suggest that these full-length 7TM C-terminal variants play important roles in mu opioid pharmacology, raising questions regarding biased signaling at these multiple C-terminal variants. In the present study, we investigated the effect of different C-terminal variants on mu agonist-induced G protein coupling, β-arrestin2 recruitment, and ultimately, signaling bias. We found that mu agonists produced marked differences in G protein activation and β-arrestin2 recruitment among various C-terminal variants, leading to biased signaling at various level. Particularly, MOR-1O, an exon 7-associated variant, showed greater β-arrestin2 bias for most mu agonists than MOR-1, an exon 4-associated variant. Biased signaling of G protein-coupled receptors has been defined by evidences that different agonists can produce divergent signaling transduction pathways through a single receptor. Our findings that a single mu agonist can induce differential signaling through multiple 7TM splice variants provide a new perspective on biased signaling at least for Oprm1, which perhaps is important for our understanding of the complex mu opioid actions in vivo where all the 7TM splice variants co-exist.
Collapse
Affiliation(s)
- Ankita Narayan
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Amanda Hunkele
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Jin Xu
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | | | - Gavril W Pasternak
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Ying-Xian Pan
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
- Department of Anesthesiology, Rutgers New Jersey Medical School, 185 South Orange Avenue, Newark, NJ, 07103, USA.
| |
Collapse
|
17
|
Weihs F, Wang J, Pfleger KDG, Dacres H. Experimental determination of the bioluminescence resonance energy transfer (BRET) Förster distances of NanoBRET and red-shifted BRET pairs. Anal Chim Acta X 2020; 6:100059. [PMID: 33392495 PMCID: PMC7772631 DOI: 10.1016/j.acax.2020.100059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/08/2020] [Accepted: 08/30/2020] [Indexed: 01/07/2023] Open
Abstract
Bioluminescence Resonance Energy Transfer (BRET) is widely applied to study protein-protein interactions, as well as increasingly to monitor both ligand binding and molecular rearrangements. The Förster distance (R0) describes the physical distance between the two chromophores at which 50% of the maximal energy transfer occurs and it depends on the choice of RET components. R0 can be experimentally determined using flexible peptide linkers of known lengths to separate the two chromophores. Knowledge of the R0 helps to inform on the choice of BRET system. For example, we have previously shown that BRET2 exhibits the largest R0 to date for any genetically encoded RET pair, which may be advantageous for investigating large macromolecular complexes if its issues of low and fast-decaying bioluminescence signal can be accommodated. In this study we have determined R0 for a range of bright and red-shifted BRET pairs, including NanoBRET with tetramethylrhodamine (TMR), non-chloro TOM (NCT), mCherry or Venus as acceptor, and BRET6, a red-shifted BRET2-like system. This study revealed R0 values of 6.15 nm and 6.94 nm for NanoBRET using TMR or NCT as acceptor ligands, respectively. R0 was 5.43 nm for NanoLuc-mCherry, 5.59 nm for NanoLuc-Venus and 5.47 nm for BRET6. This extends the palette of available BRET Förster distances, to give researchers a better-informed choice when considering BRET systems and points towards NanoBRET with NCT as a good alternative to BRET2 as an analysis tool for large macromolecular complexes. Experimental determination of Förster distances (R0) for commonly applied BRET pairs. Determination of R0 for NanoBRET with Venus, mCherry and HaloTag (TMR, NCT). Determination of R0 for BRET6. NanoLuc-HaloTag (NCT) exhibits the second largest R0 of any genetically encoded system.
Collapse
Affiliation(s)
- Felix Weihs
- CSIRO Health & Biosecurity, Parkville, 343 Royal Parade, Melbourne, VIC, 3030, Australia
| | - Jian Wang
- CSIRO Health & Biosecurity, Canberra, ACT, 2601, Australia
| | - Kevin D G Pfleger
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA, 6009, Australia.,Australian Research Council Centre for Personalised Therapeutics Technologies, Australia.,Dimerix Limited, Nedlands, WA, Australia
| | - Helen Dacres
- CSIRO Health & Biosecurity, Food Innovation Centre, 671 Sneydes Road, Werribee, VIC, 3030, Australia
| |
Collapse
|
18
|
Reichmuth AM, Zimmermann M, Wilhelm F, Frutiger A, Blickenstorfer Y, Fattinger C, Waldhoer M, Vörös J. Quantification of Molecular Interactions in Living Cells in Real Time using a Membrane Protein Nanopattern. Anal Chem 2020; 92:8983-8991. [DOI: 10.1021/acs.analchem.0c00987] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Andreas Michael Reichmuth
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, University and ETH Zurich, 8092 Zurich, Switzerland
| | | | - Florian Wilhelm
- InterAx Biotech, PARK innovAARE, 5234 Villigen, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Andreas Frutiger
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, University and ETH Zurich, 8092 Zurich, Switzerland
| | - Yves Blickenstorfer
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, University and ETH Zurich, 8092 Zurich, Switzerland
| | - Christof Fattinger
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Maria Waldhoer
- InterAx Biotech, PARK innovAARE, 5234 Villigen, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, University and ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
19
|
Brouillette RL, Besserer-Offroy É, Mona CE, Chartier M, Lavenus S, Sousbie M, Belleville K, Longpré JM, Marsault É, Grandbois M, Sarret P. Cell-penetrating pepducins targeting the neurotensin receptor type 1 relieve pain. Pharmacol Res 2020; 155:104750. [PMID: 32151680 DOI: 10.1016/j.phrs.2020.104750] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/12/2020] [Accepted: 03/05/2020] [Indexed: 01/29/2023]
Abstract
Pepducins are cell-penetrating, membrane-tethered lipopeptides designed to target the intracellular region of a G protein-coupled receptor (GPCR) in order to allosterically modulate the receptor's signaling output. In this proof-of-concept study, we explored the pain-relief potential of a pepducin series derived from the first intracellular loop of neurotensin receptor type 1 (NTS1), a class A GPCR that mediates many of the effects of the neurotensin (NT) tridecapeptide, including hypothermia, hypotension and analgesia. We used BRET-based biosensors to determine the pepducins' ability to engage G protein signaling pathways associated with NTS1 activation. We observed partial Gαq and Gα13 activation at a 10 μM concentration, indicating that these pepducins may act as allosteric agonists of NTS1. Additionally, we used surface plasmon resonance (SPR) as a label-free assay to monitor pepducin-induced responses in CHO-K1 cells stably expressing hNTS1. This whole-cell integrated assay enabled us to subdivide our pepducin series into three profile response groups. In order to determine the pepducins' antinociceptive potential, we then screened the series in an acute pain model (tail-flick test) by measuring tail withdrawal latencies to a thermal nociceptive stimulus, following intrathecal (i.t.) pepducin administration (275 nmol/kg). We further evaluated promising pepducins in a tonic pain model (formalin test), as well as in neuropathic (Chronic Constriction Injury) and inflammatory (Complete Freund's Adjuvant) chronic pain models. We report one pepducin, PP-001, that consistently reduced rat nociceptive behaviors, even in chronic pain paradigms. Finally, we designed a TAMRA-tagged version of PP-001 and found by confocal microscopy that the pepducin reached the rat dorsal root ganglia post i.t. injection, thus potentially modulating the activity of NTS1 at this location to produce its analgesic effect. Altogether, these results suggest that NTS1-derived pepducins may represent a promising strategy in pain-relief.
Collapse
Affiliation(s)
- Rebecca L Brouillette
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Élie Besserer-Offroy
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada.
| | - Christine E Mona
- Ahmanson Translational Theranostic Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
| | - Magali Chartier
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Sandrine Lavenus
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Marc Sousbie
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Karine Belleville
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Jean-Michel Longpré
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Éric Marsault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Michel Grandbois
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
20
|
CCR5: Established paradigms and new frontiers for a 'celebrity' chemokine receptor. Cytokine 2019; 109:81-93. [PMID: 29903576 DOI: 10.1016/j.cyto.2018.02.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 01/04/2023]
Abstract
Because of the level of attention it received due to its role as the principal HIV coreceptor, CCR5 has been described as a 'celebrity' chemokine receptor. Here we describe the development of CCR5 inhibitory strategies that have been developed for HIV therapy and which are now additionally being considered for use in HIV prevention and cure. The wealth of CCR5-related tools that have been developed during the intensive investigation of CCR5 as an HIV drug target can now be turned towards the study of CCR5 as a model chemokine receptor. We also summarize what is currently known about the cell biology and pharmacology of CCR5, providing an update on new areas of investigation that have emerged in recent research. Finally, we discuss the potential of CCR5 as a drug target for diseases other than HIV, discussing the evidence linking CCR5 and its natural chemokine ligands with inflammatory diseases, particularly neuroinflammation, and certain cancers. These pathologies may provide new uses for the strategies for CCR5 blockade originally developed to combat HIV/AIDS.
Collapse
|
21
|
Optical approaches for single-cell and subcellular analysis of GPCR-G protein signaling. Anal Bioanal Chem 2019; 411:4481-4508. [PMID: 30927013 DOI: 10.1007/s00216-019-01774-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 01/05/2023]
Abstract
G protein-coupled receptors (GPCRs), G proteins, and their signaling associates are major signal transducers that control the majority of cellular signaling and regulate key biological functions including immune, neurological, cardiovascular, and metabolic processes. These pathways are targeted by over one-third of drugs on the market; however, the current understanding of their function is limited and primarily derived from cell-destructive approaches providing an ensemble of static, multi-cell information about the status and composition of molecules. Spatiotemporal behavior of molecules involved is crucial to understanding in vivo cell behaviors both in health and disease, and the advent of genetically encoded fluorescence proteins and small fluorophore-based biosensors has facilitated the mapping of dynamic signaling in cells with subcellular acuity. Since we and others have developed optogenetic methods to regulate GPCR-G protein signaling in single cells and subcellular regions using dedicated wavelengths, the desire to develop and adopt optogenetically amenable assays to measure signaling has motivated us to take a broader look at the available optical tools and approaches compatible with measuring single-cell and subcellular GPCR-G protein signaling. Here we review such key optical approaches enabling the examination of GPCR, G protein, secondary messenger, and downstream molecules such as kinase and lipid signaling in living cells. The methods reviewed employ both fluorescence and bioluminescence detection. We not only further elaborate the underlying principles of these sensors but also discuss the experimental criteria and limitations to be considered during their use in single-cell and subcellular signal mapping.
Collapse
|
22
|
Dale NC, Johnstone EKM, White CW, Pfleger KDG. NanoBRET: The Bright Future of Proximity-Based Assays. Front Bioeng Biotechnol 2019; 7:56. [PMID: 30972335 PMCID: PMC6443706 DOI: 10.3389/fbioe.2019.00056] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/04/2019] [Indexed: 12/17/2022] Open
Abstract
Bioluminescence resonance energy transfer (BRET) is a biophysical technique used to monitor proximity within live cells. BRET exploits the naturally occurring phenomenon of dipole-dipole energy transfer from a donor enzyme (luciferase) to an acceptor fluorophore following enzyme-mediated oxidation of a substrate. This results in production of a quantifiable signal that denotes proximity between proteins and/or molecules tagged with complementary luciferase and fluorophore partners. BRET assays have been used to observe an array of biological functions including ligand binding, intracellular signaling, receptor-receptor proximity, and receptor trafficking, however, BRET assays can theoretically be used to monitor the proximity of any protein or molecule for which appropriate fusion constructs and/or fluorophore conjugates can be produced. Over the years, new luciferases and approaches have been developed that have increased the potential applications for BRET assays. In particular, the development of the small, bright and stable Nanoluciferase (NanoLuc; Nluc) and its use in NanoBRET has vastly broadened the potential applications of BRET assays. These advances have exciting potential to produce new experimental methods to monitor protein-protein interactions (PPIs), protein-ligand interactions, and/or molecular proximity. In addition to NanoBRET, Nluc has also been exploited to produce NanoBiT technology, which further broadens the scope of BRET to monitor biological function when NanoBiT is combined with an acceptor. BRET has proved to be a powerful tool for monitoring proximity and interaction, and these recent advances further strengthen its utility for a range of applications.
Collapse
Affiliation(s)
- Natasha C Dale
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia
| | - Elizabeth K M Johnstone
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia
| | - Carl W White
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia
| | - Kevin D G Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia.,Dimerix Limited, Nedlands, WA, Australia
| |
Collapse
|
23
|
Kobayashi H, Picard LP, Schönegge AM, Bouvier M. Bioluminescence resonance energy transfer-based imaging of protein-protein interactions in living cells. Nat Protoc 2019; 14:1084-1107. [PMID: 30911173 DOI: 10.1038/s41596-019-0129-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 01/08/2019] [Indexed: 11/09/2022]
Abstract
Bioluminescence resonance energy transfer (BRET) is a transfer of energy between a luminescence donor and a fluorescence acceptor. Because BRET occurs when the distance between the donor and acceptor is <10 nm, and its efficiency is inversely proportional to the sixth power of distance, it has gained popularity as a proximity-based assay to monitor protein-protein interactions and conformational rearrangements in live cells. In such assays, one protein of interest is fused to a bioluminescent energy donor (luciferases from Renilla reniformis or Oplophorus gracilirostris), and the other protein is fused to a fluorescent energy acceptor (such as GFP or YFP). Because the BRET donor does not require an external light source, it does not lead to phototoxicity or autofluorescence. It therefore represents an interesting alternative to fluorescence-based imaging such as FRET. However, the low signal output of BRET energy donors has limited the spatiotemporal resolution of BRET imaging. Here, we describe how recent improvements in detection devices and BRET probes can be used to markedly improve the resolution of BRET imaging, thus widening the field of BRET imaging applications. The protocol described herein involves three main stages. First, cell preparation and transfection require 3 d, including cell culture time. Second, image acquisition takes 10-120 min per sample, after an initial 60 min for microscope setup. Finally, image analysis typically takes 1-2 h. The choices of energy donor, acceptor, luminescent substrates, cameras and microscope setup, as well as acquisition modes to be used for different applications, are also discussed.
Collapse
Affiliation(s)
- Hiroyuki Kobayashi
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, Canada
| | - Louis-Philippe Picard
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, Canada
| | - Anne-Marie Schönegge
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, Canada
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
24
|
Fleiss A, Sarkisyan KS. A brief review of bioluminescent systems (2019). Curr Genet 2019; 65:877-882. [PMID: 30850867 PMCID: PMC6620254 DOI: 10.1007/s00294-019-00951-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 12/19/2022]
Abstract
Despite being widely used in reporter technologies, bioluminescent systems are largely understudied. Of at least forty different bioluminescent systems thought to exist in nature, molecular components of only seven light-emitting reactions are known, and the full biochemical pathway leading to light emission is only understood for two of them. Here, we provide a succinct overview of currently known bioluminescent systems highlighting available tools for research and discussing future applications.
Collapse
Affiliation(s)
- Aubin Fleiss
- Synthetic Biology Group, MRC London Institute of Medical Sciences, London, UK.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Karen S Sarkisyan
- Synthetic Biology Group, MRC London Institute of Medical Sciences, London, UK. .,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK. .,Planta LLC, Bolshoi Boulevard, 42 Str 1, Office 335, Moscow, 121205, Russia. .,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, Moscow, 117997, Russia.
| |
Collapse
|
25
|
El Khamlichi C, Reverchon-Assadi F, Hervouet-Coste N, Blot L, Reiter E, Morisset-Lopez S. Bioluminescence Resonance Energy Transfer as a Method to Study Protein-Protein Interactions: Application to G Protein Coupled Receptor Biology. Molecules 2019; 24:E537. [PMID: 30717191 PMCID: PMC6384791 DOI: 10.3390/molecules24030537] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/21/2019] [Accepted: 01/30/2019] [Indexed: 12/22/2022] Open
Abstract
The bioluminescence resonance energy transfer (BRET) approach involves resonance energy transfer between a light-emitting enzyme and fluorescent acceptors. The major advantage of this technique over biochemical methods is that protein-protein interactions (PPI) can be monitored without disrupting the natural environment, frequently altered by detergents and membrane preparations. Thus, it is considered as one of the most versatile technique for studying molecular interactions in living cells at "physiological" expression levels. BRET analysis has been applied to study many transmembrane receptor classes including G-protein coupled receptors (GPCR). It is well established that these receptors may function as dimeric/oligomeric forms and interact with multiple effectors to transduce the signal. Therefore, they are considered as attractive targets to identify PPI modulators. In this review, we present an overview of the different BRET systems developed up to now and their relevance to identify inhibitors/modulators of protein⁻protein interaction. Then, we introduce the different classes of agents that have been recently developed to target PPI, and provide some examples illustrating the use of BRET-based assays to identify and characterize innovative PPI modulators in the field of GPCRs biology. Finally, we discuss the main advantages and the limits of BRET approach to characterize PPI modulators.
Collapse
Affiliation(s)
- Chayma El Khamlichi
- Centre de Biophysique Moléculaire, CNRS, UPR 4301, University of Orléans and INSERM, 45071 Orléans, France.
- PRC, INRA, CNRS, Université François Rabelais-Tours, 37380 Nouzilly, France.
| | - Flora Reverchon-Assadi
- Centre de Biophysique Moléculaire, CNRS, UPR 4301, University of Orléans and INSERM, 45071 Orléans, France.
| | - Nadège Hervouet-Coste
- Centre de Biophysique Moléculaire, CNRS, UPR 4301, University of Orléans and INSERM, 45071 Orléans, France.
| | - Lauren Blot
- Centre de Biophysique Moléculaire, CNRS, UPR 4301, University of Orléans and INSERM, 45071 Orléans, France.
| | - Eric Reiter
- PRC, INRA, CNRS, Université François Rabelais-Tours, 37380 Nouzilly, France.
| | - Séverine Morisset-Lopez
- Centre de Biophysique Moléculaire, CNRS, UPR 4301, University of Orléans and INSERM, 45071 Orléans, France.
| |
Collapse
|
26
|
White CW, Johnstone EKM, See HB, Pfleger KDG. NanoBRET ligand binding at a GPCR under endogenous promotion facilitated by CRISPR/Cas9 genome editing. Cell Signal 2018; 54:27-34. [PMID: 30471466 DOI: 10.1016/j.cellsig.2018.11.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/10/2018] [Accepted: 11/20/2018] [Indexed: 01/14/2023]
Abstract
Bioluminescence resonance energy transfer (BRET) is a versatile tool used to investigate membrane receptor signalling and function. We have recently developed a homogenous NanoBRET ligand binding assay to monitor interactions between G protein-coupled receptors and fluorescent ligands. However, this assay requires the exogenous expression of a receptor fused to the nanoluciferase (Nluc) and is thus not applicable to natively-expressed receptors. To overcome this limitation in HEK293 cells, we have utilised CRISPR/Cas9 genome engineering to insert Nluc in-frame with the endogenous ADORA2B locus this resulted in HEK293 cells expressing adenosine A2B receptors under endogenous promotion tagged on their N-terminus with Nluc. As expected, we found relatively low levels of endogenous (gene-edited) Nluc/A2B receptor expression compared to cells transiently transfected with expression vectors coding for Nluc/A2B. However, in cells expressing gene-edited Nluc/A2B receptors we observed clear saturable ligand binding of a non-specific fluorescent adenosine receptor antagonist XAC-X-BY630 (Kd = 21.4 nM). Additionally, at gene-edited Nluc/A2B receptors we derived pharmacological parameters of ligand binding; Kd as well as Kon and Koff for binding of XAC-X-BY630 by NanoBRET association kinetic binding assays. Lastly, cells expressing gene-edited Nluc/A2B were used to determine the pKi of unlabelled adenosine receptor ligands in competition ligand binding assays. Utilising CRISPR/Cas9 genome engineering here we show that NanoBRET ligand binding assays can be performed at gene-edited receptors under endogenous promotion in live cells, therefore overcoming a fundamental limitation of NanoBRET ligand assays.
Collapse
Affiliation(s)
- Carl W White
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia 6009, Australia; Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia; Australian Research Council Centre for Personalised Therapeutics Technologies, Australia.
| | - Elizabeth K M Johnstone
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia 6009, Australia; Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia; Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Heng B See
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia 6009, Australia; Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia; Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Kevin D G Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia 6009, Australia; Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia; Australian Research Council Centre for Personalised Therapeutics Technologies, Australia; Dimerix Limited, Nedlands, Western Australia 6009, Australia.
| |
Collapse
|
27
|
Haider RS, Godbole A, Hoffmann C. To sense or not to sense-new insights from GPCR-based and arrestin-based biosensors. Curr Opin Cell Biol 2018; 57:16-24. [PMID: 30408632 DOI: 10.1016/j.ceb.2018.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/18/2018] [Indexed: 12/27/2022]
Abstract
Advances in resolving crystal structures of GPCRs and their binding partners as well as improvements in live-cell microscopy and the fluorescent proteins pallet has greatly driven new ideas for designing optical sensors for the same. Sensors have been developed to monitor ligand binding as well as the ensuing ligand-induced conformational changes in GPCRs, G-proteins and arrestins. In this review we will highlight the functionality of such sensor designs starting from monitoring ligand binding to receptor activation and interaction with arrestins. Furthermore, we will highlight the importance of sensor designs to monitor receptor-dependent arrestin conformations and give an idea about the various detected arrestin conformations and their possible implications.
Collapse
Affiliation(s)
- Raphael Silvanus Haider
- Institut für Molekulare Zellbiologie, CMB-Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll Straße 2, D-07745 Jena, Germany
| | - Amod Godbole
- Institut für Molekulare Zellbiologie, CMB-Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll Straße 2, D-07745 Jena, Germany
| | - Carsten Hoffmann
- Institut für Molekulare Zellbiologie, CMB-Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll Straße 2, D-07745 Jena, Germany.
| |
Collapse
|
28
|
Wang A, Feng J, Li Y, Zou P. Beyond Fluorescent Proteins: Hybrid and Bioluminescent Indicators for Imaging Neural Activities. ACS Chem Neurosci 2018; 9:639-650. [PMID: 29482322 DOI: 10.1021/acschemneuro.7b00455] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Optical biosensors have been invaluable tools in neuroscience research, as they provide the ability to directly visualize neural activity in real time, with high specificity, and with exceptional spatial and temporal resolution. Notably, a majority of these sensors are based on fluorescent protein scaffolds, which offer the ability to target specific cell types or even subcellular compartments. However, fluorescent proteins are intrinsically bulky tags, often insensitive to the environment, and always require excitation light illumination. To address these limitations, there has been a proliferation of alternative sensor scaffolds developed in recent years, including hybrid sensors that combine the advantages of synthetic fluorophores and genetically encoded protein tags, as well as bioluminescent probes. While still in their early stage of development as compared with fluorescent protein-based sensors, these novel probes have offered complementary solutions to interrogate various aspects of neuronal communication, including transmitter release, changes in membrane potential, and the production of second messengers. In this Review, we discuss these important new developments with a particular focus on design strategies.
Collapse
Affiliation(s)
- Anqi Wang
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Peng Zou
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| |
Collapse
|
29
|
Besserer-Offroy É, Bérubé P, Côté J, Murza A, Longpré JM, Dumaine R, Lesur O, Auger-Messier M, Leduc R, Marsault É, Sarret P. The hypotensive effect of activated apelin receptor is correlated with β-arrestin recruitment. Pharmacol Res 2018. [PMID: 29530600 DOI: 10.1016/j.phrs.2018.02.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The apelinergic system is an important player in the regulation of both vascular tone and cardiovascular function, making this physiological system an attractive target for drug development for hypertension, heart failure and ischemic heart disease. Indeed, apelin exerts a positive inotropic effect in humans whilst reducing peripheral vascular resistance. In this study, we investigated the signaling pathways through which apelin exerts its hypotensive action. We synthesized a series of apelin-13 analogs whereby the C-terminal Phe13 residue was replaced by natural or unnatural amino acids. In HEK293 cells expressing APJ, we evaluated the relative efficacy of these compounds to activate Gαi1 and GαoA G-proteins, recruit β-arrestins 1 and 2 (βarrs), and inhibit cAMP production. Calculating the transduction ratio for each pathway allowed us to identify several analogs with distinct signaling profiles. Furthermore, we found that these analogs delivered i.v. to Sprague-Dawley rats exerted a wide range of hypotensive responses. Indeed, two compounds lost their ability to lower blood pressure, while other analogs significantly reduced blood pressure as apelin-13. Interestingly, analogs that did not lower blood pressure were less effective at recruiting βarrs. Finally, using Spearman correlations, we established that the hypotensive response was significantly correlated with βarr recruitment but not with G protein-dependent signaling. In conclusion, our results demonstrated that the βarr recruitment potency is involved in the hypotensive efficacy of activated APJ.
Collapse
Affiliation(s)
- Élie Besserer-Offroy
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Patrick Bérubé
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Jérôme Côté
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Alexandre Murza
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Jean-Michel Longpré
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Robert Dumaine
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Olivier Lesur
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Mannix Auger-Messier
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Éric Marsault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| |
Collapse
|
30
|
Brox R, Milanos L, Saleh N, Baumeister P, Buschauer A, Hofmann D, Heinrich MR, Clark T, Tschammer N. Molecular Mechanisms of Biased and Probe-Dependent Signaling at CXC-Motif Chemokine Receptor CXCR3 Induced by Negative Allosteric Modulators. Mol Pharmacol 2018; 93:309-322. [PMID: 29343553 DOI: 10.1124/mol.117.110296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 01/12/2018] [Indexed: 12/21/2022] Open
Abstract
Our recent explorations of allosteric modulators with improved properties resulted in the identification of two biased negative allosteric modulators, BD103 (N-1-{[3-(4-ethoxyphenyl)-4-oxo-3,4-dihydropyrido[2,3-d]pyrimi-din2yl]ethyl}-4-(4-fluorobutoxy)-N-[(1-methylpiperidin-4-yl)methyl}]butanamide) and BD064 (5-[(N-{1-[3-(4-ethoxyphenyl)-4-oxo-3,4-dihydropyrido[2,3-d]pyrimidin-2-yl]ethyl-2-[4-fluoro-3-(trifluoromethyl)phenyl]acetamido)methyl]-2-fluorophenyl}boronic acid), that exhibited probe-dependent inhibition of CXC-motif chemokine receptor CXCR3 signaling. With the intention to elucidate the structural mechanisms underlying their selectivity and probe dependence, we used site-directed mutagenesis combined with homology modeling and docking to identify amino acids of CXCR3 that contribute to modulator binding, signaling, and transmission of cooperativity. With the use of allosteric radioligand RAMX3 ([3H]N-{1-[3-(4-ethoxyphenyl)-4-oxo-3,4-dihydropyrido[2,3-d]pyrimidin-2-yl]ethyl}-2-[4-fluoro-3-(trifluoromethyl)phenyl]-N-[(1-methylpiperidin-4-yl)methyl]acetamide), we identified that F1313.32 and Y3087.43 contribute specifically to the binding pocket of BD064, whereas D1864.60 solely participates in the stabilization of binding conformation of BD103. The influence of mutations on the ability of negative allosteric modulators to inhibit chemokine-mediated activation (CXCL11 and CXCL10) was assessed with the bioluminescence resonance energy transfer-based cAMP and β-arrestin recruitment assay. Obtained data revealed complex molecular mechanisms governing biased and probe-dependent signaling at CXCR3. In particular, F1313.32, S3047.39, and Y3087.43 emerged as key residues for the compounds to modulate the chemokine response. Notably, D1864.60, W2686.48, and S3047.39 turned out to play a role in signal pathway selectivity of CXCL10, as mutations of these residues led to a G protein-active but β-arrestin-inactive conformation. These diverse effects of mutations suggest the existence of ligand- and pathway-specific receptor conformations and give new insights in the sophisticated signaling machinery between allosteric ligands, chemokines, and their receptors, which can provide a powerful platform for the development of new allosteric drugs with improved pharmacological properties.
Collapse
Affiliation(s)
- Regine Brox
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center (R.B., D.H., M.R.H., N.T.) and Computer Chemistry Center (L.M., N.S., T.C.), Friedrich Alexander University, Erlangen, Germany; and Institute of Pharmacy, University of Regensburg, Regensburg, Germany (P.B., A.B.)
| | - Lampros Milanos
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center (R.B., D.H., M.R.H., N.T.) and Computer Chemistry Center (L.M., N.S., T.C.), Friedrich Alexander University, Erlangen, Germany; and Institute of Pharmacy, University of Regensburg, Regensburg, Germany (P.B., A.B.)
| | - Noureldin Saleh
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center (R.B., D.H., M.R.H., N.T.) and Computer Chemistry Center (L.M., N.S., T.C.), Friedrich Alexander University, Erlangen, Germany; and Institute of Pharmacy, University of Regensburg, Regensburg, Germany (P.B., A.B.)
| | - Paul Baumeister
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center (R.B., D.H., M.R.H., N.T.) and Computer Chemistry Center (L.M., N.S., T.C.), Friedrich Alexander University, Erlangen, Germany; and Institute of Pharmacy, University of Regensburg, Regensburg, Germany (P.B., A.B.)
| | - Armin Buschauer
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center (R.B., D.H., M.R.H., N.T.) and Computer Chemistry Center (L.M., N.S., T.C.), Friedrich Alexander University, Erlangen, Germany; and Institute of Pharmacy, University of Regensburg, Regensburg, Germany (P.B., A.B.)
| | - Dagmar Hofmann
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center (R.B., D.H., M.R.H., N.T.) and Computer Chemistry Center (L.M., N.S., T.C.), Friedrich Alexander University, Erlangen, Germany; and Institute of Pharmacy, University of Regensburg, Regensburg, Germany (P.B., A.B.)
| | - Markus R Heinrich
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center (R.B., D.H., M.R.H., N.T.) and Computer Chemistry Center (L.M., N.S., T.C.), Friedrich Alexander University, Erlangen, Germany; and Institute of Pharmacy, University of Regensburg, Regensburg, Germany (P.B., A.B.)
| | - Timothy Clark
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center (R.B., D.H., M.R.H., N.T.) and Computer Chemistry Center (L.M., N.S., T.C.), Friedrich Alexander University, Erlangen, Germany; and Institute of Pharmacy, University of Regensburg, Regensburg, Germany (P.B., A.B.)
| | - Nuska Tschammer
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center (R.B., D.H., M.R.H., N.T.) and Computer Chemistry Center (L.M., N.S., T.C.), Friedrich Alexander University, Erlangen, Germany; and Institute of Pharmacy, University of Regensburg, Regensburg, Germany (P.B., A.B.)
| |
Collapse
|
31
|
Visualization of ligand-induced dopamine D 2S and D 2L receptor internalization by TIRF microscopy. Sci Rep 2017; 7:10894. [PMID: 28883522 PMCID: PMC5589927 DOI: 10.1038/s41598-017-11436-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/23/2017] [Indexed: 01/11/2023] Open
Abstract
G protein-coupled receptors (GPCRs), including the dopamine receptors, represent a group of important pharmacological targets. Upon agonist binding, GPCRs frequently undergo internalization, a process that is known to attenuate functional responses upon prolonged exposure to agonists. In this study, internalization was visualized by means of total internal reflection fluorescence (TIRF) microscopy at a level of discrete single events near the plasma membrane with high spatial resolution. A novel method has been developed to determine the relative extent of internalized fluorescent receptor-ligand complexes by comparative fluorescence quantification in living CHO cells. The procedure entails treatment with the reducing agent sodium borohydride, which converts cyanine-based fluorescent ligands on the membrane surface to a long-lived reduced form. Because the highly polar reducing agent is not able to pass the cell membrane, the fluorescent receptor-ligand complexes located in internalized compartments remain fluorescent under TIRF illumination. We applied the method to investigate differences of the short (D2S) and the long (D2L) isoforms of dopamine D2 receptors in their ability to undergo agonist-induced internalization.
Collapse
|
32
|
Wacker D, Wang S, McCorvy JD, Betz RM, Venkatakrishnan AJ, Levit A, Lansu K, Schools ZL, Che T, Nichols DE, Shoichet BK, Dror RO, Roth BL. Crystal Structure of an LSD-Bound Human Serotonin Receptor. Cell 2017; 168:377-389.e12. [PMID: 28129538 PMCID: PMC5289311 DOI: 10.1016/j.cell.2016.12.033] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/29/2016] [Accepted: 12/21/2016] [Indexed: 12/20/2022]
Abstract
The prototypical hallucinogen LSD acts via serotonin receptors, and here we describe the crystal structure of LSD in complex with the human serotonin receptor 5-HT2B. The complex reveals conformational rearrangements to accommodate LSD, providing a structural explanation for the conformational selectivity of LSD's key diethylamide moiety. LSD dissociates exceptionally slow from both 5-HT2BR and 5-HT2AR-a major target for its psychoactivity. Molecular dynamics (MD) simulations suggest that LSD's slow binding kinetics may be due to a "lid" formed by extracellular loop 2 (EL2) at the entrance to the binding pocket. A mutation predicted to increase the mobility of this lid greatly accelerates LSD's binding kinetics and selectively dampens LSD-mediated β-arrestin2 recruitment. This study thus reveals an unexpected binding mode of LSD; illuminates key features of its kinetics, stereochemistry, and signaling; and provides a molecular explanation for LSD's actions at human serotonin receptors. PAPERCLIP.
Collapse
Affiliation(s)
- Daniel Wacker
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599-7365, USA.
| | - Sheng Wang
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599-7365, USA
| | - John D McCorvy
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599-7365, USA
| | - Robin M Betz
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA 94305, USA; Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - A J Venkatakrishnan
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Anat Levit
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158-2280, USA
| | - Katherine Lansu
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599-7365, USA
| | - Zachary L Schools
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599-7365, USA
| | - Tao Che
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599-7365, USA
| | - David E Nichols
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7360, USA
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158-2280, USA
| | - Ron O Dror
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA 94305, USA; Biophysics Program, Stanford University, Stanford, CA 94305, USA.
| | - Bryan L Roth
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599-7365, USA; Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7360, USA; National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), School of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599-7365, USA.
| |
Collapse
|
33
|
Characterizing Dynamic Protein-Protein Interactions Using the Genetically Encoded Split Biosensor Assay Technique Split TEV. Methods Mol Biol 2017; 1596:219-238. [PMID: 28293890 DOI: 10.1007/978-1-4939-6940-1_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dynamic protein-protein interactions (PPIs) are fundamental building blocks of cellular signaling and monitoring their regulation promotes the understanding of signaling in health and disease. Genetically encoded split protein biosensor assays, such as the split TEV method, have proved to be highly valuable when studying regulated PPIs in living cells. Split TEV is based on the functional complementation of two previously inactive TEV protease fragments fused to interacting proteins and provides a robust, sensitive and flexible readout to monitor PPIs both at the membrane and in the cytosol. Thus, split TEV can be used to analyze interactomes of receptors, membrane-associated proteins, and cytosolic proteins. In particular, split TEV is useful to assay activities of relevant drug targets, such as receptor tyrosine kinases and G protein-coupled receptors, in compound screens. As split TEV uses genetically encoded readouts, including standard reporters based on fluorescence and luminescence, the technique can also be combined with scalable molecular barcode reporter systems, allowing the integration into multiplexed high-throughput assay approaches. Split TEV can be used in standard heterologous cell lines and primary cell types, including neurons, either in a transient or stably integrated format. When using cell lines, the basic protocol takes 30-96 h to complete, depending on the complexity of the experimental question addressed.
Collapse
|
34
|
Foster SR, Bräuner-Osborne H. Investigating Internalization and Intracellular Trafficking of GPCRs: New Techniques and Real-Time Experimental Approaches. Handb Exp Pharmacol 2017; 245:41-61. [PMID: 29018878 DOI: 10.1007/164_2017_57] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The ability to regulate the interaction between cells and their extracellular environment is essential for the maintenance of appropriate physiological function. For G protein-coupled receptors (GPCRs), this regulation occurs through multiple mechanisms that provide spatial and temporal control for signal transduction. One of the major mechanisms for GPCR regulation involves their endocytic trafficking, which serves to internalize the receptors from the plasma membrane and thereby attenuate G protein-dependent signaling. However, there is accumulating evidence to suggest that GPCRs can signal independently of G proteins, as well as from intracellular compartments including endosomes. It is in this context that receptor internalization and intracellular trafficking have attracted renewed interest within the GPCR field. In this chapter, we will review the current understanding and methodologies that have been used to investigate internalization and intracellular signaling of GPCRs, with a particular focus on emerging real-time techniques. These recent developments have improved our understanding of the complexities of GPCR internalization and intracellular signaling and suggest that the broader biological relevance and potential therapeutic implications of these processes remain to be explored.
Collapse
Affiliation(s)
- Simon R Foster
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark.
| |
Collapse
|
35
|
Berchiche YA, Sakmar TP. CXC Chemokine Receptor 3 Alternative Splice Variants Selectively Activate Different Signaling Pathways. Mol Pharmacol 2016; 90:483-95. [PMID: 27512119 DOI: 10.1124/mol.116.105502] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/09/2016] [Indexed: 01/17/2023] Open
Abstract
The G protein-coupled receptor (GPCR) C-X-C chemokine receptor 3 (CXCR3) is a potential drug target that mediates signaling involved in cancer metastasis and inflammatory diseases. The CXCR3 primary transcript has three potential alternative splice variants and cell-type specific expression results in receptor variants that are believed to have different functional characteristics. However, the molecular pharmacology of ligand binding to CXCR3 alternative splice variants and their downstream signaling pathways remain poorly explored. To better understand the role of the functional consequences of alternative splicing of CXCR3, we measured signaling in response to four different chemokine ligands (CXCL4, CXCL9, CXCL10, and CXCL11) with agonist activity at CXCR3. Both CXCL10 and CXCL11 activated splice variant CXCR3A. Whereas CXCL10 displayed full agonistic activity for Gαi activation and extracellular signal regulated kinase (ERK) 1/2 phosphorylation and partial agonist activity for β-arrestin recruitment, CXCL9 triggered only modest ERK1/2 phosphorylation. CXCL11 induced CXCR3B-mediated β-arrestin recruitment and little ERK phosphorylation. CXCR3Alt signaling was limited to modest ligand-induced receptor internalization and ERK1/2 phosphorylation in response to chemokines CXCL11, CXCL10, and CXCL9. These results show that CXCR3 splice variants activate different signaling pathways and that CXCR3 variant function is not redundant, suggesting a mechanism for tissue specific biased agonism. Our data show an additional layer of complexity for chemokine receptor signaling that might be exploited to target specific CXCR3 splice variants.
Collapse
Affiliation(s)
- Yamina A Berchiche
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (Y.A.B.; T.P.S.); and Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Huddinge, Sweden (T.P.S)
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (Y.A.B.; T.P.S.); and Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Huddinge, Sweden (T.P.S)
| |
Collapse
|
36
|
Tian H, Fürstenberg A, Huber T. Labeling and Single-Molecule Methods To Monitor G Protein-Coupled Receptor Dynamics. Chem Rev 2016; 117:186-245. [DOI: 10.1021/acs.chemrev.6b00084] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- He Tian
- Laboratory of Chemical Biology
and Signal Transduction, The Rockefeller University, 1230 York
Avenue, New York, New York 10065, United States
| | - Alexandre Fürstenberg
- Laboratory of Chemical Biology
and Signal Transduction, The Rockefeller University, 1230 York
Avenue, New York, New York 10065, United States
| | - Thomas Huber
- Laboratory of Chemical Biology
and Signal Transduction, The Rockefeller University, 1230 York
Avenue, New York, New York 10065, United States
| |
Collapse
|
37
|
Galandrin S, Onfroy L, Poirot MC, Sénard JM, Galés C. Delineating biased ligand efficacy at 7TM receptors from an experimental perspective. Int J Biochem Cell Biol 2016; 77:251-63. [PMID: 27107932 DOI: 10.1016/j.biocel.2016.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/15/2016] [Accepted: 04/16/2016] [Indexed: 12/17/2022]
Abstract
During the last 10 years, the concept of "biased agonism" also called "functional selectivity" swamped the pharmacology of 7 transmembrane receptors and paved the way for developing signaling pathway-selective drugs with increased efficacy and less adverse effects. Initially thought to select the activation of only a subset of the signaling pathways by the reference agonist, bias ligands revealed higher complexity as they have been shown to stabilize variable receptor conformations that associate with distinct signaling events from the reference. Today, one major challenge relies on the in vitro determination of the bias and classification of these ligands, as a prerequisite for future in vivo and clinical translation. In this review, current experimental considerations for the bias evaluation related to choice of the cellular model, of the signaling pathway as well as of the assays are presented and discussed.
Collapse
Affiliation(s)
- Ségolène Galandrin
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université Toulouse, F-31432 Toulouse, France
| | - Lauriane Onfroy
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université Toulouse, F-31432 Toulouse, France
| | - Mathias Charles Poirot
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université Toulouse, F-31432 Toulouse, France
| | - Jean-Michel Sénard
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université Toulouse, F-31432 Toulouse, France; Service de Pharmacologie Clinique, Faculté de médecine, Centre Hospitalier Universitaire de Toulouse, Université de Toulouse, F-31000 Toulouse, France
| | - Céline Galés
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université Toulouse, F-31432 Toulouse, France.
| |
Collapse
|
38
|
|
39
|
Mihara K, Ramachandran R, Saifeddine M, Hansen KK, Renaux B, Polley D, Gibson S, Vanderboor C, Hollenberg MD. Thrombin-Mediated Direct Activation of Proteinase-Activated Receptor-2: Another Target for Thrombin Signaling. Mol Pharmacol 2016; 89:606-14. [PMID: 26957205 DOI: 10.1124/mol.115.102723] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 03/03/2016] [Indexed: 01/30/2023] Open
Abstract
Thrombin is known to signal to cells by cleaving/activating a G-protein-coupled family of proteinase-activated receptors (PARs). The signaling mechanism involves the proteolytic unmasking of an N-terminal receptor sequence that acts as a tethered receptor-activating ligand. To date, the recognized targets of thrombin cleavage and activation for signaling are PAR1 and PAR4, in which thrombin cleaves at a conserved target arginine to reveal a tethered ligand. PAR2, which like PAR1 is also cleaved at an N-terminal arginine to unmask its tethered ligand, is generally regarded as a target for trypsin but not for thrombin signaling. We now show that thrombin, at concentrations that can be achieved at sites of acute injury or in a tumor microenvironment, can directly activate PAR2 vasorelaxation and signaling, stimulating calcium and mitogen-activated protein kinase responses along with triggeringβ-arrestin recruitment. Thus, PAR2 can be added alongside PAR1 and PAR4 to the targets, whereby thrombin can affect tissue function.
Collapse
Affiliation(s)
- Koichiro Mihara
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology and Pharmacology (K.M., R.R., M.S., K.K.H., B.R., D.P., S.G., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (C.V., R.R.)
| | - Rithwik Ramachandran
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology and Pharmacology (K.M., R.R., M.S., K.K.H., B.R., D.P., S.G., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (C.V., R.R.)
| | - Mahmoud Saifeddine
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology and Pharmacology (K.M., R.R., M.S., K.K.H., B.R., D.P., S.G., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (C.V., R.R.)
| | - Kristina K Hansen
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology and Pharmacology (K.M., R.R., M.S., K.K.H., B.R., D.P., S.G., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (C.V., R.R.)
| | - Bernard Renaux
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology and Pharmacology (K.M., R.R., M.S., K.K.H., B.R., D.P., S.G., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (C.V., R.R.)
| | - Danny Polley
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology and Pharmacology (K.M., R.R., M.S., K.K.H., B.R., D.P., S.G., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (C.V., R.R.)
| | - Stacy Gibson
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology and Pharmacology (K.M., R.R., M.S., K.K.H., B.R., D.P., S.G., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (C.V., R.R.)
| | - Christina Vanderboor
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology and Pharmacology (K.M., R.R., M.S., K.K.H., B.R., D.P., S.G., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (C.V., R.R.)
| | - Morley D Hollenberg
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology and Pharmacology (K.M., R.R., M.S., K.K.H., B.R., D.P., S.G., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (C.V., R.R.)
| |
Collapse
|
40
|
Coriano C, Powell E, Xu W. Monitoring Ligand-Activated Protein-Protein Interactions Using Bioluminescent Resonance Energy Transfer (BRET) Assay. Methods Mol Biol 2016; 1473:3-15. [PMID: 27518618 DOI: 10.1007/978-1-4939-6346-1_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The bioluminescent resonance energy transfer (BRET) assay has been extensively used in cell-based and in vivo imaging systems for detecting protein-protein interactions in the native environment of living cells. These protein-protein interactions are essential for the functional response of many signaling pathways to environmental chemicals. BRET has been used as a toxicological tool for identifying chemicals that either induce or inhibit these protein-protein interactions. This chapter focuses on describing the toxicological applications of BRET and its optimization as a high-throughput detection system in live cells. Here we review the construction of BRET fusion proteins, describe the BRET methodology, and outline strategies to overcome obstacles that may arise. Furthermore, we describe the advantage of BRET over other resonance energy transfer methods for monitoring protein-protein interactions.
Collapse
Affiliation(s)
- Carlos Coriano
- Department of Oncology, University of Wisconsin-Madison, 7459 WIMR II, 1111 Highland Avenue, Madison, WI, 53705-2275, USA
| | - Emily Powell
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wei Xu
- Department of Oncology, University of Wisconsin-Madison, 7459 WIMR II, 1111 Highland Avenue, Madison, WI, 53705-2275, USA.
| |
Collapse
|
41
|
Vacchini A, Busnelli M, Chini B, Locati M, Borroni EM. Analysis of G Protein and β-Arrestin Activation in Chemokine Receptors Signaling. Methods Enzymol 2016; 570:421-40. [DOI: 10.1016/bs.mie.2015.09.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
42
|
Analysis of Arrestin Recruitment to Chemokine Receptors by Bioluminescence Resonance Energy Transfer. Methods Enzymol 2016; 570:131-53. [DOI: 10.1016/bs.mie.2015.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
43
|
Robertson DN, Sleno R, Nagi K, Pétrin D, Hébert TE, Pineyro G. Design and construction of conformational biosensors to monitor ion channel activation: A prototype FlAsH/BRET-approach to Kir3 channels. Methods 2016. [DOI: 10.1016/j.ymeth.2015.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
44
|
Mo XL, Luo Y, Ivanov AA, Su R, Havel JJ, Li Z, Khuri FR, Du Y, Fu H. Enabling systematic interrogation of protein-protein interactions in live cells with a versatile ultra-high-throughput biosensor platform. J Mol Cell Biol 2015; 8:271-81. [PMID: 26578655 DOI: 10.1093/jmcb/mjv064] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/09/2015] [Indexed: 01/07/2023] Open
Abstract
Large-scale genomics studies have generated vast resources for in-depth understanding of vital biological and pathological processes. A rising challenge is to leverage such enormous information to rapidly decipher the intricate protein-protein interactions (PPIs) for functional characterization and therapeutic interventions. While a number of powerful technologies have been employed to detect PPIs, a singular PPI biosensor platform with both high sensitivity and robustness in a mammalian cell environment remains to be established. Here we describe the development and integration of a highly sensitive NanoLuc luciferase-based bioluminescence resonance energy transfer technology, termed BRET(n), which enables ultra-high-throughput (uHTS) PPI detection in live cells with streamlined co-expression of biosensors in a miniaturized format. We further demonstrate the application of BRET(n) in uHTS format in chemical biology research, including the discovery of chemical probes that disrupt PRAS40 dimerization and pathway connectivity profiling among core members of the Hippo signaling pathway. Such hippo pathway profiling not only confirmed previously reported PPIs, but also revealed two novel interactions, suggesting new mechanisms for regulation of Hippo signaling. Our BRET(n) biosensor platform with uHTS capability is expected to accelerate systematic PPI network mapping and PPI modulator-based drug discovery.
Collapse
Affiliation(s)
- Xiu-Lei Mo
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yin Luo
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Andrei A Ivanov
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rina Su
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA Department of Dermatology, XiangYa Hospital, Central South University, Changsha 410008, China
| | - Jonathan J Havel
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zenggang Li
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Fadlo R Khuri
- Department of Hematology and Medical Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Yuhong Du
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Haian Fu
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA Department of Hematology and Medical Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
45
|
Abstract
Modulation of protein-protein interactions (PPIs) is becoming increasingly important in drug discovery and chemical biology. While a few years ago this 'target class' was deemed to be largely undruggable an impressing number of publications and success stories now show that targeting PPIs with small, drug-like molecules indeed is a feasible approach. Here, we summarize the current state of small-molecule inhibition and stabilization of PPIs and review the active molecules from a structural and medicinal chemistry angle, especially focusing on the key examples of iNOS, LFA-1 and 14-3-3.
Collapse
|
46
|
Donthamsetti P, Quejada JR, Javitch JA, Gurevich VV, Lambert NA. Using Bioluminescence Resonance Energy Transfer (BRET) to Characterize Agonist-Induced Arrestin Recruitment to Modified and Unmodified G Protein-Coupled Receptors. ACTA ACUST UNITED AC 2015; 70:2.14.1-2.14.14. [PMID: 26331887 DOI: 10.1002/0471141755.ph0214s70] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
G protein-coupled receptors (GPCRs) represent ∼25% of current drug targets. Ligand binding to these receptors activates G proteins and arrestins, which are involved in differential signaling pathways. Because functionally selective or biased ligands activate one of these two pathways, they may be superior medications for certain diseases states. The identification of such ligands requires robust drug screening assays for both G protein and arrestin activity. This unit describes protocols for two bioluminescence resonance energy transfer (BRET)-based assays used to monitor arrestin recruitment to GPCRs. One assay requires modification of GPCRs by fusion to a BRET donor or acceptor moiety, whereas the other can detect arrestin recruitment to unmodified GPCRs.
Collapse
Affiliation(s)
- Prashant Donthamsetti
- Departments of Psychiatry and Pharmacology, Columbia University College of Physicians and Surgeons, New York, New York.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York
| | - Jose Rafael Quejada
- Departments of Psychiatry and Pharmacology, Columbia University College of Physicians and Surgeons, New York, New York.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York
| | - Jonathan A Javitch
- Departments of Psychiatry and Pharmacology, Columbia University College of Physicians and Surgeons, New York, New York.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York
| | | | - Nevin A Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
47
|
Chen Y, Tang H, Seibel W, Papoian R, Li X, Lambert NA, Palczewski K. A High-Throughput Drug Screening Strategy for Detecting Rhodopsin P23H Mutant Rescue and Degradation. Invest Ophthalmol Vis Sci 2015; 56:2553-67. [PMID: 25783607 DOI: 10.1167/iovs.14-16298] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Inherent instability of the P23H mutant opsin accounts for approximately 10% of autosomal dominant retinitis pigmentosa cases. Our purpose was to develop an overall set of reliable screening strategies to assess if either stabilization or enhanced degradation of mutant rhodopsin could rescue rod photoreceptors expressing this mutant protein. These strategies promise to reveal active compounds and clarify molecular mechanisms of biologically important processes, such as inhibition of target degradation or enhanced target folding. METHODS Cell-based bioluminescence reporter assays were developed and validated for high-throughput screening (HTS) of compounds that promote either stabilization or degradation of P23H mutant opsin. Such assays were further complemented by immunoblotting and image-based analyses. RESULTS Two stabilization assays of P23H mutant opsin were developed and validated, one based on β-galactosidase complementarity and a second assay involving bioluminescence resonance energy transfer (BRET) technology. Moreover, two additional assays evaluating mutant protein degradation also were employed, one based on the disappearance of luminescence and another employing the ALPHA immunoassay. Imaging of cells revealed the cellular localization of mutant rhodopsin, whereas immunoblots identified changes in the aggregation and glycosylation of P23H mutant opsin. CONCLUSIONS Our findings indicate that these initial HTS and following assays can identify active therapeutic compounds, even for difficult targets such as mutant rhodopsin. The assays are readily scalable and their function has been proven with model compounds. High-throughput screening, supported by automated imaging and classic immunoassays, can further characterize multiple steps and pathways in the biosynthesis and degradation of this essential visual system protein.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Department of Pharmacology Case Western Reserve University, Cleveland, Ohio, United States
| | - Hong Tang
- Drug Discovery Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States
| | - William Seibel
- Drug Discovery Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States
| | - Ruben Papoian
- Drug Discovery Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States
| | - Xiaoyu Li
- Department of Pharmacology Case Western Reserve University, Cleveland, Ohio, United States
| | - Nevin A Lambert
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, Georgia, United States
| | - Krzysztof Palczewski
- Department of Pharmacology Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
48
|
Machleidt T, Woodroofe CC, Schwinn MK, Méndez J, Robers MB, Zimmerman K, Otto P, Daniels DL, Kirkland TA, Wood KV. NanoBRET--A Novel BRET Platform for the Analysis of Protein-Protein Interactions. ACS Chem Biol 2015; 10:1797-804. [PMID: 26006698 DOI: 10.1021/acschembio.5b00143] [Citation(s) in RCA: 341] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dynamic interactions between proteins comprise a key mechanism for temporal control of cellular function and thus hold promise for development of novel drug therapies. It remains technically challenging, however, to quantitatively characterize these interactions within the biologically relevant context of living cells. Although, bioluminescence resonance energy transfer (BRET) has often been used for this purpose, its general applicability has been hindered by limited sensitivity and dynamic range. We have addressed this by combining an extremely bright luciferase (Nanoluc) with a means for tagging intracellular proteins with a long-wavelength fluorophore (HaloTag). The small size (19 kDa), high emission intensity, and relatively narrow spectrum (460 nm peak intensity) make Nanoluc luciferase well suited as an energy donor. By selecting an efficient red-emitting fluorophore (635 nm peak intensity) for attachment onto the HaloTag, an overall spectral separation exceeding 175 nm was achieved. This combination of greater light intensity with improved spectral resolution results in substantially increased detection sensitivity and dynamic range over current BRET technologies. Enhanced performance is demonstrated using several established model systems, as well as the ability to image BRET in individual cells. The capabilities are further exhibited in a novel assay developed for analyzing the interactions of bromodomain proteins with chromatin in living cells.
Collapse
Affiliation(s)
- Thomas Machleidt
- Promega Corporation, 2800 Woods
Hollow, Madison, Wisconsin 53711, United States
| | - Carolyn C. Woodroofe
- Promega Biosciences Incorporated, 277 Granada Drive, San Luis Obispo, California 93401, United States
| | - Marie K. Schwinn
- Promega Corporation, 2800 Woods
Hollow, Madison, Wisconsin 53711, United States
| | - Jacqui Méndez
- Promega Corporation, 2800 Woods
Hollow, Madison, Wisconsin 53711, United States
| | - Matthew B. Robers
- Promega Corporation, 2800 Woods
Hollow, Madison, Wisconsin 53711, United States
| | - Kris Zimmerman
- Promega Corporation, 2800 Woods
Hollow, Madison, Wisconsin 53711, United States
| | - Paul Otto
- Promega Corporation, 2800 Woods
Hollow, Madison, Wisconsin 53711, United States
| | - Danette L. Daniels
- Promega Corporation, 2800 Woods
Hollow, Madison, Wisconsin 53711, United States
| | - Thomas A. Kirkland
- Promega Biosciences Incorporated, 277 Granada Drive, San Luis Obispo, California 93401, United States
| | - Keith V. Wood
- Promega Corporation, 2800 Woods
Hollow, Madison, Wisconsin 53711, United States
| |
Collapse
|
49
|
Brown NE, Blumer JB, Hepler JR. Bioluminescence resonance energy transfer to detect protein-protein interactions in live cells. Methods Mol Biol 2015; 1278:457-465. [PMID: 25859969 PMCID: PMC4682348 DOI: 10.1007/978-1-4939-2425-7_30] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Bioluminescence resonance energy transfer (BRET) is a valuable tool to detect protein-protein interactions. BRET utilizes bioluminescent and fluorescent protein tags with compatible emission and excitation properties, making it possible to examine resonance energy transfer when the tags are in close proximity (<10 nm) as a typical result of protein-protein interactions. Here we describe a protocol for detecting BRET from two known protein binding partners (Gαi1 and RGS14) in HEK 293 cells using Renilla luciferase and yellow fluorescent protein tags. We discuss the calculation of the acceptor/donor ratio as well as net BRET and demonstrate that BRET can be used as a platform to investigate the regulation of protein-protein interactions in live cells in real time.
Collapse
Affiliation(s)
- Nicole E Brown
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | | |
Collapse
|
50
|
Levoye A, Zwier JM, Jaracz-Ros A, Klipfel L, Cottet M, Maurel D, Bdioui S, Balabanian K, Prézeau L, Trinquet E, Durroux T, Bachelerie F. A Broad G Protein-Coupled Receptor Internalization Assay that Combines SNAP-Tag Labeling, Diffusion-Enhanced Resonance Energy Transfer, and a Highly Emissive Terbium Cryptate. Front Endocrinol (Lausanne) 2015; 6:167. [PMID: 26617570 PMCID: PMC4638144 DOI: 10.3389/fendo.2015.00167] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 10/19/2015] [Indexed: 12/15/2022] Open
Abstract
Although G protein-coupled receptor (GPCR) internalization has long been considered as a major aspect of the desensitization process that tunes ligand responsiveness, internalization is also involved in receptor resensitization and signaling, as well as the ligand scavenging function of some atypical receptors. Internalization thus contributes to the diversity of GPCR-dependent signaling, and its dynamics and quantification in living cells has generated considerable interest. We developed a robust and sensitive assay to follow and quantify ligand-induced and constitutive-induced GPCR internalization but also receptor recycling in living cells. This assay is based on diffusion-enhanced resonance energy transfer (DERET) between cell surface GPCRs labeled with a luminescent terbium cryptate donor and a fluorescein acceptor present in the culture medium. GPCR internalization results in a quantifiable reduction of energy transfer. This method yields a high signal-to-noise ratio due to time-resolved measurements. For various GPCRs belonging to different classes, we demonstrated that constitutive and ligand-induced internalization could be monitored as a function of time and ligand concentration, thus allowing accurate quantitative determination of kinetics of receptor internalization but also half-maximal effective or inhibitory concentrations of compounds. In addition to its selectivity and sensitivity, we provided evidence that DERET-based internalization assay is particularly suitable for characterizing biased ligands. Furthermore, the determination of a Z'-factor value of 0.45 indicates the quality and suitability of DERET-based internalization assay for high-throughput screening (HTS) of compounds that may modulate GPCRs internalization.
Collapse
Affiliation(s)
- Angélique Levoye
- INSERM U1148, Laboratory of Vascular Translational Science, Université Paris 13, Sorbonne Paris Cité, Paris, France
- *Correspondence: Angélique Levoye,
| | | | - Agnieszka Jaracz-Ros
- INSERM UMR996, Inflammation, Chemokines and Immunopathology, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Laurence Klipfel
- INSERM UMR996, Inflammation, Chemokines and Immunopathology, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Martin Cottet
- CNRS UMR 5203, INSERM U1191, Institut de Génomique Fonctionnelle, Université Montpellier 1 & 2, Montpellier, France
| | - Damien Maurel
- CNRS UMR 5203, INSERM U1191, Institut de Génomique Fonctionnelle, Université Montpellier 1 & 2, Montpellier, France
| | | | - Karl Balabanian
- INSERM UMR996, Inflammation, Chemokines and Immunopathology, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Laurent Prézeau
- CNRS UMR 5203, INSERM U1191, Institut de Génomique Fonctionnelle, Université Montpellier 1 & 2, Montpellier, France
| | | | - Thierry Durroux
- CNRS UMR 5203, INSERM U1191, Institut de Génomique Fonctionnelle, Université Montpellier 1 & 2, Montpellier, France
| | - Françoise Bachelerie
- INSERM UMR996, Inflammation, Chemokines and Immunopathology, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| |
Collapse
|