151
|
Vischer HF, Castro M, Pin JP. G Protein-Coupled Receptor Multimers: A Question Still Open Despite the Use of Novel Approaches. Mol Pharmacol 2015; 88:561-71. [PMID: 26138074 DOI: 10.1124/mol.115.099440] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 07/02/2015] [Indexed: 12/11/2022] Open
Abstract
Heteromerization of G protein-coupled receptors (GPCRs) can significantly change the functional properties of involved receptors. Various biochemical and biophysical methodologies have been developed in the last two decades to identify and functionally evaluate GPCR heteromers in heterologous cells, with recent approaches focusing on GPCR complex stoichiometry and stability. Yet validation of these observations in native tissues is still lagging behind for the majority of GPCR heteromers. Remarkably, recent studies, particularly some involving advanced fluorescence microscopy techniques, are contributing to our current knowledge of aspects that were not well known until now, such as GPCR complex stoichiometry and stability. In parallel, a growing effort is being applied to move the field forward into native systems. This short review will highlight recent developments to study the stoichiometry and stability of GPCR complexes and methodologies to detect native GPCR dimers.
Collapse
Affiliation(s)
- Henry F Vischer
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, Amsterdam, The Netherlands (H.F.V.); Molecular Pharmacology Laboratory, Biofarma Research Group (GI-1685), University of Santiago de Compostela, Center for Research in Molecular Medicine and Chronic Diseases, Santiago de Compostela, Spain (M.C.); and Centre National de la Recherche Scientifique, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France (J.-P.P.)
| | - Marián Castro
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, Amsterdam, The Netherlands (H.F.V.); Molecular Pharmacology Laboratory, Biofarma Research Group (GI-1685), University of Santiago de Compostela, Center for Research in Molecular Medicine and Chronic Diseases, Santiago de Compostela, Spain (M.C.); and Centre National de la Recherche Scientifique, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France (J.-P.P.)
| | - Jean-Philippe Pin
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, Amsterdam, The Netherlands (H.F.V.); Molecular Pharmacology Laboratory, Biofarma Research Group (GI-1685), University of Santiago de Compostela, Center for Research in Molecular Medicine and Chronic Diseases, Santiago de Compostela, Spain (M.C.); and Centre National de la Recherche Scientifique, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France (J.-P.P.)
| |
Collapse
|
152
|
LI G, SANCHEZ V, NAGARAJ P, KHAN S, RAJPOOT N. A novel multitarget tracking algorithm for Myosin VI protein molecules on actin filaments in TIRFM sequences. J Microsc 2015; 260:312-25. [DOI: 10.1111/jmi.12299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 07/09/2015] [Indexed: 11/28/2022]
Affiliation(s)
- G. LI
- Department of Computer Science; University of Warwick; UK
| | - V. SANCHEZ
- Department of Computer Science; University of Warwick; UK
| | | | - S. KHAN
- Department of Biology, Lahore University of Management Sciences (LUMS); Pakistan
| | - N. RAJPOOT
- Department of Computer Science; University of Warwick; UK
- Department of Computer Science & Engineering (CSE); Qatar University; Qatar
| |
Collapse
|
153
|
Abstract
Adrenergic signaling, in particular signaling in the sympathetic nervous system, is a prime example of the control of an essential physiological system. It has served as a model system both for the control of mediator release and for receptor signaling and regulation. This review covers the historical development of the field and then addresses issues that represent key fields of ongoing research: the mechanisms and kinetics of receptor activation, temporal patterns of downstream signaling and signal bias, receptor mobility and aggregation, and signal compartmentation and specificity. The available evidence suggests that adrenergic signaling may involve complex spatiotemporal patterns, which give texture to the signaling process and may contain additional biological information.
Collapse
Affiliation(s)
- Martin J Lohse
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078, Würzburg, Germany.
- Rudolf Virchow Center, University of Würzburg, Versbacher Str. 9, 97078, Würzburg, Germany.
- Comprehensive Heart Failure Center, University of Würzburg, Versbacher Str. 9, 97078, Würzburg, Germany.
| |
Collapse
|
154
|
Cheng M, Zhang W, Yuan J, Luo W, Li N, Lin S, Yang Y, Fang X, Chen PR. Single-molecule dynamics of site-specific labeled transforming growth factor type II receptors on living cells. Chem Commun (Camb) 2015; 50:14724-7. [PMID: 24887482 DOI: 10.1039/c4cc02804j] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We achieved single-molecule imaging and tracking of the transforming growth factor type II receptor (TβRII) that was labeled by an organic dye via a genetically encoded unnatural amino acid (UAA) and the copper-free click chemistry. The stoichiometry, mobility and dimerization kinetics of individual TβRII molecules were determined.
Collapse
Affiliation(s)
- Ming Cheng
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Key Laboratory of Molecular Nanostructures and Nanotechnology, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Marsango S, Caltabiano G, Pou C, Varela Liste MJ, Milligan G. Analysis of Human Dopamine D3 Receptor Quaternary Structure. J Biol Chem 2015; 290:15146-62. [PMID: 25931118 PMCID: PMC4463457 DOI: 10.1074/jbc.m114.630681] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 04/23/2015] [Indexed: 01/11/2023] Open
Abstract
The dopamine D3 receptor is a class A, rhodopsin-like G protein-coupled receptor that can form dimers and/or higher order oligomers. However, the molecular basis for production of these complexes is not well defined. Using combinations of molecular modeling, site-directed mutagenesis, and homogenous time-resolved FRET, the interfaces that allow dopamine D3 receptor monomers to interact were defined and used to describe likely quaternary arrangements of the receptor. These were then compared with published crystal structures of dimeric β1-adrenoreceptor, μ-opioid, and CXCR4 receptors. The data indicate important contributions of residues from within each of transmembrane domains I, II, IV, V, VI, and VII as well as the intracellular helix VIII in the formation of D3-D3 receptor interfaces within homo-oligomers and are consistent with the D3 receptor adopting a β1-adrenoreceptor-like quaternary arrangement. Specifically, results suggest that D3 protomers can interact with each other via at least two distinct interfaces: the first one comprising residues from transmembrane domains I and II along with those from helix VIII and a second one involving transmembrane domains IV and V. Moreover, rather than existing only as distinct dimeric species, the results are consistent with the D3 receptor also assuming a quaternary structure in which two transmembrane domain I-II-helix VIII dimers interact to form a "rhombic" tetramer via an interface involving residues from transmembrane domains VI and VII. In addition, the results also provide insights into the potential contribution of molecules of cholesterol to the overall organization and potential stability of the D3 receptor and possibly other GPCR quaternary structures.
Collapse
Affiliation(s)
- Sara Marsango
- From the Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom and
| | - Gianluigi Caltabiano
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Chantevy Pou
- From the Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom and
| | - María José Varela Liste
- From the Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom and
| | - Graeme Milligan
- From the Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom and
| |
Collapse
|
156
|
Liste MJV, Caltabiano G, Ward RJ, Alvarez-Curto E, Marsango S, Milligan G. The molecular basis of oligomeric organization of the human M3 muscarinic acetylcholine receptor. Mol Pharmacol 2015; 87:936-53. [PMID: 25769304 DOI: 10.1124/mol.114.096925] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/13/2015] [Indexed: 11/22/2022] Open
Abstract
G protein-coupled receptors, including the M3 muscarinic acetylcholine receptor, can form homo-oligomers. However, the basis of these interactions and the overall organizational structure of such oligomers are poorly understood. Combinations of site-directed mutagenesis and homogenous time-resolved fluorescence resonance energy transfer studies that assessed interactions between receptor protomers at the surface of transfected cells indicated important contributions of regions of transmembrane domains I, IV, V, VI, and VII as well as intracellular helix VIII to the overall organization. Molecular modeling studies based on both these results and an X-ray structure of the inactive state of the M3 receptor bound by the antagonist/inverse agonist tiotropium were then employed. The results could be accommodated fully by models in which a proportion of the cell surface M3 receptor population is a tetramer with rhombic, but not linear, orientation. This is consistent with previous studies based on spectrally resolved, multiphoton fluorescence resonance energy transfer. Modeling studies furthermore suggest an important role for molecules of cholesterol at the dimer + dimer interface of the tetramer, which is consistent with the presence of cholesterol at key locations in many G protein-coupled receptor crystal structures. Mutants that displayed disrupted quaternary organization were often poorly expressed and showed immature N-glycosylation. Sustained treatment of cells expressing such mutants with the muscarinic receptor inverse agonist atropine increased cellular levels and restored both cell surface delivery and quaternary organization to many of the mutants. These observations suggest that organization as a tetramer may occur before plasma membrane delivery and may be a key step in cellular quality control assessment.
Collapse
Affiliation(s)
- María José Varela Liste
- Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom (M.J.V.L., G.C., R.J.W., E.A.-C., S.M., G.M.), and Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain (G.C.)
| | - Gianluigi Caltabiano
- Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom (M.J.V.L., G.C., R.J.W., E.A.-C., S.M., G.M.), and Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain (G.C.)
| | - Richard J Ward
- Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom (M.J.V.L., G.C., R.J.W., E.A.-C., S.M., G.M.), and Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain (G.C.)
| | - Elisa Alvarez-Curto
- Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom (M.J.V.L., G.C., R.J.W., E.A.-C., S.M., G.M.), and Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain (G.C.)
| | - Sara Marsango
- Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom (M.J.V.L., G.C., R.J.W., E.A.-C., S.M., G.M.), and Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain (G.C.)
| | - Graeme Milligan
- Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom (M.J.V.L., G.C., R.J.W., E.A.-C., S.M., G.M.), and Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain (G.C.)
| |
Collapse
|
157
|
Stoddart LA, Kilpatrick LE, Briddon SJ, Hill SJ. Probing the pharmacology of G protein-coupled receptors with fluorescent ligands. Neuropharmacology 2015; 98:48-57. [PMID: 25979488 DOI: 10.1016/j.neuropharm.2015.04.033] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/23/2015] [Accepted: 04/29/2015] [Indexed: 01/01/2023]
Abstract
G protein-coupled receptors control a wide range of physiological processes and are the target for many clinically used drugs. Understanding the way in which receptors bind agonists and antagonists, their organisation in the membrane and their regulation after agonist binding are important properties which are key to developing new drugs. One way to achieve this knowledge is through the use of fluorescent ligands, which have been used to study the expression and function of receptors in endogenously expressing systems. Fluorescent ligands with appropriate imaging properties can be used in conjunction with confocal microscopy to investigate the regulation of receptors after activation. Alternatively, through the use of single molecule microscopy, they can probe the spatial organisation of receptors within the membrane. This review focuses on the techniques in which fluorescent ligands have been used and the novel aspects of G protein-coupled receptor pharmacology which have been uncovered. This article is part of the Special Issue entitled 'Fluorescent Tools in Neuropharmacology'.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Laura E Kilpatrick
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Stephen J Briddon
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Stephen J Hill
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
158
|
Langosch D, Scharnagl C, Steiner H, Lemberg MK. Understanding intramembrane proteolysis: from protein dynamics to reaction kinetics. Trends Biochem Sci 2015; 40:318-27. [PMID: 25941170 DOI: 10.1016/j.tibs.2015.04.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 12/12/2022]
Abstract
Intramembrane proteolysis - cleavage of proteins within the plane of a membrane - is a widespread phenomenon that can contribute to the functional activation of substrates and is involved in several diseases. Although different families of intramembrane proteases have been discovered and characterized, we currently do not know how these enzymes discriminate between substrates and non-substrates, how site-specific cleavage is achieved, or which factors determine the rate of proteolysis. Focusing on γ-secretase and rhomboid proteases, we argue that answers to these questions may emerge from connecting experimental readouts, such as reaction kinetics and the determination of cleavage sites, to the structures and the conformational dynamics of substrates and enzymes.
Collapse
Affiliation(s)
- D Langosch
- Technische Universität München, Lehrstuhl Chemie der Biopolymere, Weihenstephaner Berg 3, 85354 Freising, and Munich Center for Integrated Protein Science (CIMPS(M)), Germany.
| | - C Scharnagl
- Fakultät für Physik E14, Technische Universität München, Maximus-von-Imhof-Forum 4, 85354 Freising, Germany
| | - H Steiner
- Ludwig-Maximilians-University Munich, Metabolic Biochemistry and DZNE (German Center for Neurodegenerative Diseases), Munich, Germany
| | - M K Lemberg
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Deutsches Krebsforschungszentrum (DKFZ)-ZMBH Allianz, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| |
Collapse
|
159
|
Fang Y. Total internal reflection fluorescence quantification of receptor pharmacology. BIOSENSORS 2015; 5:223-240. [PMID: 25922915 PMCID: PMC4493547 DOI: 10.3390/bios5020223] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 04/15/2015] [Accepted: 04/17/2015] [Indexed: 12/30/2022]
Abstract
Total internal reflection fluorescence (TIRF) microscopy has been widely used as a single molecule imaging technique to study various fundamental aspects of cell biology, owing to its ability to selectively excite a very thin fluorescent volume immediately above the substrate on which the cells are grown. However, TIRF microscopy has found little use in high content screening due to its complexity in instrumental setup and experimental procedures. Inspired by the recent demonstration of label-free evanescent wave biosensors for cell phenotypic profiling and drug screening with high throughput, we had hypothesized and demonstrated that TIRF imaging is also amenable to receptor pharmacology profiling. This paper reviews key considerations and recent applications of TIRF imaging for pharmacology profiling.
Collapse
Affiliation(s)
- Ye Fang
- Biochemical Technologies, Science and Technology Division, Corning Incorporated, Corning, NY 14831, USA.
| |
Collapse
|
160
|
Aslanoglou D, Alvarez-Curto E, Marsango S, Milligan G. Distinct Agonist Regulation of Muscarinic Acetylcholine M2-M3 Heteromers and Their Corresponding Homomers. J Biol Chem 2015; 290:14785-96. [PMID: 25918156 PMCID: PMC4505543 DOI: 10.1074/jbc.m115.649079] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Indexed: 01/01/2023] Open
Abstract
Each subtype of the muscarinic receptor family of G protein-coupled receptors is activated by similar concentrations of the neurotransmitter acetylcholine or closely related synthetic analogs such as carbachol. However, pharmacological selectivity can be generated by the introduction of a pair of mutations to produce Receptor Activated Solely by Synthetic Ligand (RASSL) forms of muscarinic receptors. These display loss of potency for acetylcholine/carbachol alongside a concurrent gain in potency for the ligand clozapine N-oxide. Co-expression of a form of wild type human M2 and a RASSL variant of the human M3 receptor resulted in concurrent detection of each of M2-M2 and M3-M3 homomers alongside M2-M3 heteromers at the surface of stably transfected Flp-InTM T-RExTM 293 cells. In this setting occupancy of the receptors with a muscarinic antagonist was without detectable effect on any of the muscarinic oligomers. However, selective agonist occupancy of the M2 receptor resulted in enhanced M2-M2 homomer interactions but decreased M2-M3 heteromer interactions. By contrast, selective activation of the M3 RASSL receptor did not significantly alter either M3-M3 homomer or M2-M3 heteromer interactions. Selectively targeting closely related receptor oligomers may provide novel therapeutic opportunities.
Collapse
Affiliation(s)
- Despoina Aslanoglou
- From the Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Elisa Alvarez-Curto
- From the Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Sara Marsango
- From the Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Graeme Milligan
- From the Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| |
Collapse
|
161
|
Ward RJ, Pediani JD, Godin AG, Milligan G. Regulation of oligomeric organization of the serotonin 5-hydroxytryptamine 2C (5-HT2C) receptor observed by spatial intensity distribution analysis. J Biol Chem 2015; 290:12844-57. [PMID: 25825490 PMCID: PMC4432300 DOI: 10.1074/jbc.m115.644724] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Indexed: 12/19/2022] Open
Abstract
The questions of whether G protein-coupled receptors exist as monomers, dimers, and/or oligomers and if these species interconvert in a ligand-dependent manner are among the most contentious current issues in biology. When employing spatial intensity distribution analysis to laser scanning confocal microscope images of cells stably expressing either a plasma membrane-associated form of monomeric enhanced green fluorescent protein (eGFP) or a tandem version of this fluorophore, the eGFP tandem was identified as a dimer. Similar studies on cells stably expressing an eGFP-tagged form of the epidermal growth factor receptor demonstrated that, although largely a monomer in the basal state, this receptor rapidly became predominantly dimeric upon the addition of its ligand epidermal growth factor. In cells induced to express an eGFP-tagged form of the serotonin 5-hydroxytryptamine 2C (5-HT2C) receptor, global analysis of construct quantal brightness was consistent with the predominant form of the receptor being dimeric. However, detailed spatial intensity distribution analysis demonstrated the presence of multiple forms ranging from monomers to higher-order oligomers. Furthermore, treatment with chemically distinct 5-HT2C receptor antagonists resulted in a time-dependent change in the quaternary organization to one in which there was a preponderance of receptor monomers. This antagonist-mediated effect was reversible, because washout of the ligand resulted in the regeneration of many of the oligomeric forms of the receptor.
Collapse
Affiliation(s)
- Richard J Ward
- From the Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - John D Pediani
- From the Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Antoine G Godin
- the University of Bordeaux, LP2N, UMR 5298, F-33405 Talence, France, and the Institut d'Optique Graduate School and CNRS, LP2N, UMR 5298, F-33405 Talence, France
| | - Graeme Milligan
- From the Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom,
| |
Collapse
|
162
|
Mashanov GI. Single molecule dynamics in a virtual cell: a three-dimensional model that produces simulated fluorescence video-imaging data. J R Soc Interface 2015; 11:20140442. [PMID: 25008080 PMCID: PMC4233692 DOI: 10.1098/rsif.2014.0442] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The analysis of single molecule imaging experiments is complicated by the stochastic nature of single molecule events, by instrument noise and by the limited information which can be gathered about any individual molecule observed. Consequently, it is important to cross check experimental results using a model simulating single molecule dynamics (e.g. movements and binding events) in a virtual cell-like environment. The output of such a model should match the real data format allowing researchers to compare simulated results with the real experiments. The proposed model exploits the advantages of ‘object-oriented’ computing. First of all, the ability to create and manipulate a number of classes, each containing an arbitrary number of single molecule objects. These classes may include objects moving within the ‘cytoplasm’; objects moving at the ‘plasma membrane’; and static objects located inside the ‘body’. The objects of a given class can interact with each other and/or with the objects of other classes according to their physical and chemical properties. Each model run generates a sequence of images, each containing summed images of all fluorescent objects emitting light under given illumination conditions with realistic levels of noise and emission fluctuations. The model accurately reproduces reported single molecule experiments and predicts the outcome of future experiments.
Collapse
Affiliation(s)
- Gregory I Mashanov
- Division of Physical Biochemistry, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| |
Collapse
|
163
|
Bellot M, Galandrin S, Boularan C, Matthies HJ, Despas F, Denis C, Javitch J, Mazères S, Sanni SJ, Pons V, Seguelas MH, Hansen JL, Pathak A, Galli A, Sénard JM, Galés C. Dual agonist occupancy of AT1-R-α2C-AR heterodimers results in atypical Gs-PKA signaling. Nat Chem Biol 2015; 11:271-9. [PMID: 25706338 DOI: 10.1038/nchembio.1766] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/09/2015] [Indexed: 12/25/2022]
Abstract
Hypersecretion of norepinephrine (NE) and angiotensin II (AngII) is a hallmark of major prevalent cardiovascular diseases that contribute to cardiac pathophysiology and morbidity. Herein, we explore whether heterodimerization of presynaptic AngII AT1 receptor (AT1-R) and NE α2C-adrenergic receptor (α2C-AR) could underlie their functional cross-talk to control NE secretion. Multiple bioluminescence resonance energy transfer and protein complementation assays allowed us to accurately probe the structures and functions of the α2C-AR-AT1-R dimer promoted by ligand binding to individual protomers. We found that dual agonist occupancy resulted in a conformation of the heterodimer different from that induced by active individual protomers and triggered atypical Gs-cAMP-PKA signaling. This specific pharmacological signaling unit was identified in vivo to promote not only NE hypersecretion in sympathetic neurons but also sympathetic hyperactivity in mice. Thus, we uncovered a new process by which GPCR heterodimerization creates an original functional pharmacological entity and that could constitute a promising new target in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Morgane Bellot
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Ségolène Galandrin
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Cédric Boularan
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Heinrich J Matthies
- 1] Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA. [2] Neuroscience Program in Substance Abuse, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Fabien Despas
- 1] Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Toulouse, France. [2] Service de Pharmacologie Clinique, Centre Hospitalier Universitaire de Toulouse, Faculté de Médecine, Toulouse, France
| | - Colette Denis
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Jonathan Javitch
- 1] Center for Molecular Recognition and Departments of Psychiatry and Pharmacology, Columbia University, New York, New York, USA. [2] College of Physicians and Surgeons, Columbia University, New York, New York, USA. [3] Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Serge Mazères
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS, UMR 508, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Samra Joke Sanni
- 1] Department of Clinical Biochemistry, Glostrup Hospital, Glostrup, Denmark. [2] Diabetes Biology and Metabolism, Novo Nordisk, Måløv, Denmark
| | - Véronique Pons
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Marie-Hélène Seguelas
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Jakob L Hansen
- Diabetes Biology and Metabolism, Novo Nordisk, Måløv, Denmark
| | - Atul Pathak
- 1] Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Toulouse, France. [2] Service de Pharmacologie Clinique, Centre Hospitalier Universitaire de Toulouse, Faculté de Médecine, Toulouse, France
| | - Aurelio Galli
- 1] Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA. [2] Neuroscience Program in Substance Abuse, Vanderbilt University School of Medicine, Nashville, Tennessee, USA. [3] Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, USA. [4] Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jean-Michel Sénard
- 1] Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Toulouse, France. [2] Service de Pharmacologie Clinique, Centre Hospitalier Universitaire de Toulouse, Faculté de Médecine, Toulouse, France
| | - Céline Galés
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
164
|
Yano Y, Kondo K, Kitani R, Yamamoto A, Matsuzaki K. Cholesterol-Induced Lipophobic Interaction between Transmembrane Helices Using Ensemble and Single-Molecule Fluorescence Resonance Energy Transfer. Biochemistry 2015; 54:1371-9. [DOI: 10.1021/bi501528e] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Yoshiaki Yano
- Graduate School of Pharmaceutical
Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kotaro Kondo
- Graduate School of Pharmaceutical
Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Ryota Kitani
- Graduate School of Pharmaceutical
Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Arisa Yamamoto
- Graduate School of Pharmaceutical
Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Katsumi Matsuzaki
- Graduate School of Pharmaceutical
Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
165
|
Affiliation(s)
- Nevin A Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA, 30912-2300, USA
| | - Jonathan A Javitch
- Departments of Psychiatry and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| |
Collapse
|
166
|
Bouvier M, Hébert TE. CrossTalk proposal: Weighing the evidence for Class A GPCR dimers, the evidence favours dimers. J Physiol 2015; 592:2439-41. [PMID: 24931944 DOI: 10.1113/jphysiol.2014.272252] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Michel Bouvier
- Département de Biochimie, Institut de Recherch en Immunologie and Cancérologie (IRIC), Université de Montréal, Montréal, Québec, Canada, H3T 1J4
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada, H3G 1Y6
| |
Collapse
|
167
|
Lambert NA, Javitch JA. CrossTalk opposing view: Weighing the evidence for class A GPCR dimers, the jury is still out. J Physiol 2015; 592:2443-5. [PMID: 24931945 DOI: 10.1113/jphysiol.2014.272997] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Nevin A Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA, 30912-2300, USA
| | - Jonathan A Javitch
- Departments of Psychiatry and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| |
Collapse
|
168
|
Xue L, Rovira X, Scholler P, Zhao H, Liu J, Pin JP, Rondard P. Major ligand-induced rearrangement of the heptahelical domain interface in a GPCR dimer. Nat Chem Biol 2015; 11:134-40. [PMID: 25503927 DOI: 10.1038/nchembio.1711] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 10/14/2014] [Indexed: 12/30/2022]
Abstract
G protein-coupled receptors (GPCRs) are major players in cell communication. Although they form functional monomers, increasing evidence indicates that GPCR dimerization has a critical role in cooperative phenomena that are important for cell signal integration. However, the structural bases of these phenomena remain elusive. Here, using well-characterized receptor dimers, the metabotropic glutamate receptors (mGluRs), we show that structural changes at the dimer interface are linked to receptor activation. We demonstrate that the main dimer interface is formed by transmembrane α helix 4 (TM4) and TM5 in the inactive state and by TM6 in the active state. This major change in the dimer interface is required for receptor activity because locking the TM4-TM5 interface prevents activation by agonist, whereas locking the TM6 interface leads to a constitutively active receptor. These data provide important information on the activation mechanism of mGluRs and improve our understanding of the structural basis of the negative cooperativity observed in these GPCR dimers.
Collapse
MESH Headings
- Alanine/genetics
- Animals
- COS Cells
- Chlorocebus aethiops
- Cysteine/genetics
- HEK293 Cells
- Humans
- Ligands
- Models, Molecular
- Mutagenesis, Site-Directed
- Protein Binding
- Protein Multimerization
- Protein Structure, Tertiary
- Protein Subunits
- Rats
- Receptor, Metabotropic Glutamate 5/agonists
- Receptor, Metabotropic Glutamate 5/chemistry
- Receptor, Metabotropic Glutamate 5/genetics
- Receptors, GABA-B/chemistry
- Receptors, GABA-B/genetics
- Receptors, GABA-B/metabolism
- Receptors, Metabotropic Glutamate/agonists
- Receptors, Metabotropic Glutamate/chemistry
- Receptors, Metabotropic Glutamate/genetics
- Transcriptional Activation
- Transfection
Collapse
Affiliation(s)
- Li Xue
- 1] Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China. [2] CNRS, UMR5203, Institut de Génomique Fonctionnelle, Montpellier, France. [3] INSERM, U661, Montpellier, France. [4] Université Montpellier 1 &2, Montpellier, France
| | - Xavier Rovira
- 1] CNRS, UMR5203, Institut de Génomique Fonctionnelle, Montpellier, France. [2] INSERM, U661, Montpellier, France. [3] Université Montpellier 1 &2, Montpellier, France
| | - Pauline Scholler
- 1] CNRS, UMR5203, Institut de Génomique Fonctionnelle, Montpellier, France. [2] INSERM, U661, Montpellier, France. [3] Université Montpellier 1 &2, Montpellier, France
| | - Han Zhao
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jianfeng Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jean-Philippe Pin
- 1] CNRS, UMR5203, Institut de Génomique Fonctionnelle, Montpellier, France. [2] INSERM, U661, Montpellier, France. [3] Université Montpellier 1 &2, Montpellier, France
| | - Philippe Rondard
- 1] CNRS, UMR5203, Institut de Génomique Fonctionnelle, Montpellier, France. [2] INSERM, U661, Montpellier, France. [3] Université Montpellier 1 &2, Montpellier, France
| |
Collapse
|
169
|
Suzuki KG. New Insights into the Organization of Plasma Membrane and Its Role in Signal Transduction. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 317:67-96. [DOI: 10.1016/bs.ircmb.2015.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
170
|
Abstract
The publication of high-resolution structures for all of the opioid receptor subfamilies has unveiled exciting opportunities for mechanistic insight into the molecular mechanisms underlying the biology of nociception, reward, and higher cognitive functions, as well as promises for progress in several clinical areas such as pain management, physiological dependence, addiction, and mood disorders. To turn this promise into novel and improved therapeutic entities, however, this information needs to be supplemented with research strategies that explore the dynamic behavior of the proteins and their interactions with other receptors and ligands in their physiological environment.Here we describe state-of-the-art molecular dynamics computational protocols, based on all-atom and coarse-grained modeling techniques, designed to estimate crucial thermodynamic and kinetic parameters describing the binding of small-molecule ligands and the formation of supramolecular complexes.
Collapse
|
171
|
Liu L, Woolf A, Rodriguez AW, Capasso F. Absolute position total internal reflection microscopy with an optical tweezer. Proc Natl Acad Sci U S A 2014; 111:E5609-15. [PMID: 25512542 PMCID: PMC4284537 DOI: 10.1073/pnas.1422178112] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A noninvasive, in situ calibration method for total internal reflection microscopy (TIRM) based on optical tweezing is presented, which greatly expands the capabilities of this technique. We show that by making only simple modifications to the basic TIRM sensing setup and procedure, a probe particle's absolute position relative to a dielectric interface may be known with better than 10 nm precision out to a distance greater than 1 μm from the surface. This represents an approximate 10× improvement in error and 3× improvement in measurement range over conventional TIRM methods. The technique's advantage is in the direct measurement of the probe particle's scattering intensity vs. height profile in situ, rather than relying on assumptions, inexact system analogs, or detailed knowledge of system parameters for calibration. To demonstrate the improved versatility of the TIRM method in terms of tunability, precision, and range, we show our results for the hindered near-wall diffusion coefficient for a spherical dielectric particle.
Collapse
Affiliation(s)
- Lulu Liu
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138; and
| | - Alexander Woolf
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138; and
| | | | - Federico Capasso
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138; and
| |
Collapse
|
172
|
Fricke F, Dietz MS, Heilemann M. Single-Molecule Methods to Study Membrane Receptor Oligomerization. Chemphyschem 2014; 16:713-21. [DOI: 10.1002/cphc.201402765] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Indexed: 11/06/2022]
|
173
|
Jonas KC, Fanelli F, Huhtaniemi IT, Hanyaloglu AC. Single molecule analysis of functionally asymmetric G protein-coupled receptor (GPCR) oligomers reveals diverse spatial and structural assemblies. J Biol Chem 2014; 290:3875-92. [PMID: 25516594 PMCID: PMC4326798 DOI: 10.1074/jbc.m114.622498] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Formation of G protein-coupled receptors (GPCRs) into dimers and higher order oligomers represents a key mechanism in pleiotropic signaling, yet how individual protomers function within oligomers remains poorly understood. We present a super-resolution imaging approach, resolving single GPCR molecules to ∼8 nm resolution in functional asymmetric dimers and oligomers using dual-color photoactivatable dyes and localization microscopy (PD-PALM). PD-PALM of two functionally defined mutant luteinizing hormone receptors (LHRs), a ligand-binding deficient receptor (LHRB−) and a signaling-deficient (LHRS−) receptor, which only function via intermolecular cooperation, favored oligomeric over dimeric formation. PD-PALM imaging of trimers and tetramers revealed specific spatial organizations of individual protomers in complexes where the ratiometric composition of LHRB− to LHRS− modulated ligand-induced signal sensitivity. Structural modeling of asymmetric LHR oligomers strongly aligned with PD-PALM-imaged spatial arrangements, identifying multiple possible helix interfaces mediating inter-protomer associations. Our findings reveal that diverse spatial and structural assemblies mediating GPCR oligomerization may acutely fine-tune the cellular signaling profile.
Collapse
Affiliation(s)
- Kim C Jonas
- From the Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Francesca Fanelli
- the Computational Structural Biology Lab, Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 183-41100 Modena, Italy, and
| | - Ilpo T Huhtaniemi
- From the Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom, the Institute for Biomedicine, Department of Physiology, University of Turku, 20520 Turku, Finland
| | - Aylin C Hanyaloglu
- From the Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom,
| |
Collapse
|
174
|
Adamson RJ, Watts A. Kinetics of the early events of GPCR signalling. FEBS Lett 2014; 588:4701-7. [PMID: 25447525 PMCID: PMC4266533 DOI: 10.1016/j.febslet.2014.10.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 10/10/2014] [Accepted: 10/27/2014] [Indexed: 11/28/2022]
Abstract
Little is known of the kinetics of interactions between GPCRs and their signalling partners. NTS1 binds Gαi1 and Gαs with affinities of 15 ± 6 nM and 31 ± 18 nM (SE), respectively. This SPR assay may be applicable to multiple partners in the signalling cascade. We provide the first direct evidence for GPCR-G protein coupling in nanodiscs.
Neurotensin receptor type 1 (NTS1) is a G protein-coupled receptor (GPCR) that affects cellular responses by initiating a cascade of interactions through G proteins. The kinetic details for these interactions are not well-known. Here, NTS1-nanodisc-Gαs and Gαi1 interactions were studied. The binding affinities of Gαi1 and Gαs to NTS1 were directly measured by surface plasmon resonance (SPR) and determined to be 15 ± 6 nM and 31 ± 18 nM, respectively. This SPR configuration permits the kinetics of early events in signalling pathways to be explored and can be used to initiate descriptions of the GPCR interactome.
Collapse
Affiliation(s)
- Roslin J Adamson
- Biomembrane Structure Unit, Biochemistry Department, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Anthony Watts
- Biomembrane Structure Unit, Biochemistry Department, University of Oxford, South Parks Road, Oxford OX1 3QU, UK.
| |
Collapse
|
175
|
Guidolin D, Agnati LF, Marcoli M, Borroto-Escuela DO, Fuxe K. G-protein-coupled receptor type A heteromers as an emerging therapeutic target. Expert Opin Ther Targets 2014; 19:265-83. [PMID: 25381716 DOI: 10.1517/14728222.2014.981155] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The discovery of receptor-receptor interactions (RRIs) in the early 1980s provided evidence that G-protein-coupled receptors (GPCRs) operate not only as monomers but also as heteromers, in which integration of the incoming signals takes place already at the plasma membrane level through allosteric RRIs. These integrative mechanisms give sophisticated dynamics to the structure and function of these receptor assemblies in terms of modulation of recognition, G-protein signaling and selectivity and switching to β-arrestin signaling. AREAS COVERED The present review briefly describes the concept of direct RRI and the available data on the mechanisms of oligomer formation. Further, pharmacological data concerning the best characterized heteromers involving type A GPCRs will be analyzed to evaluate their profile as possible targets for the treatment of various diseases, in particular of impacting diseases of the CNS. EXPERT OPINION GPCR heteromers have the potential to open a completely new field for pharmacology with likely a major impact in molecular medicine. Novel pharmacological strategies for the treatment of several pathologies have already been proposed. However, several challenges still exist to accurately characterize the role of the identified heteroreceptor complexes in pathology and to develop heteromer-specific ligands capable of efficiently exploiting their pharmacological features.
Collapse
Affiliation(s)
- Diego Guidolin
- University of Padova, Department of Molecular Medicine , via Gabelli 65, 35121 Padova , Italy +39 049 8272316 ; +39 049 8272319 ;
| | | | | | | | | |
Collapse
|
176
|
Pi J, Jin H, Yang F, Chen ZW, Cai J. In situ single molecule imaging of cell membranes: linking basic nanotechniques to cell biology, immunology and medicine. NANOSCALE 2014; 6:12229-12249. [PMID: 25227707 DOI: 10.1039/c4nr04195j] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The cell membrane, which consists of a viscous phospholipid bilayer, different kinds of proteins and various nano/micrometer-sized domains, plays a very important role in ensuring the stability of the intracellular environment and the order of cellular signal transductions. Exploring the precise cell membrane structure and detailed functions of the biomolecules in a cell membrane would be helpful to understand the underlying mechanisms involved in cell membrane signal transductions, which could further benefit research into cell biology, immunology and medicine. The detection of membrane biomolecules at the single molecule level can provide some subtle information about the molecular structure and the functions of the cell membrane. In particular, information obtained about the molecular mechanisms and other information at the single molecule level are significantly different from that detected from a large amount of biomolecules at the large-scale through traditional techniques, and can thus provide a novel perspective for the study of cell membrane structures and functions. However, the precise investigations of membrane biomolecules prompts researchers to explore cell membranes at the single molecule level by the use of in situ imaging methods, as the exact conformation and functions of biomolecules are highly controlled by the native cellular environment. Recently, the in situ single molecule imaging of cell membranes has attracted increasing attention from cell biologists and immunologists. The size of biomolecules and their clusters on the cell surface are set at the nanoscale, which makes it mandatory to use high- and super-resolution imaging techniques to realize the in situ single molecule imaging of cell membranes. In the past few decades, some amazing imaging techniques and instruments with super resolution have been widely developed for molecule imaging, which can also be further employed for the in situ single molecule imaging of cell membranes. In this review, we attempt to summarize the characteristics of these advanced techniques for use in the in situ single molecule imaging of cell membranes. We believe that this work will help to promote the technological and methodological developments of super-resolution techniques for the single molecule imaging of cell membranes and help researchers better understand which technique is most suitable for their future exploring of membrane biomolecules; ultimately promoting further developments in cell biology, immunology and medicine.
Collapse
Affiliation(s)
- Jiang Pi
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technique, Macau, China.
| | | | | | | | | |
Collapse
|
177
|
Vernall AJ, Hill SJ, Kellam B. The evolving small-molecule fluorescent-conjugate toolbox for Class A GPCRs. Br J Pharmacol 2014; 171:1073-84. [PMID: 23734587 DOI: 10.1111/bph.12265] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 05/09/2013] [Accepted: 05/17/2013] [Indexed: 01/15/2023] Open
Abstract
The past decade has witnessed fluorescently tagged drug molecules gaining significant attraction in their use as pharmacological tools with which to visualize and interrogate receptor targets at the single-cell level. Additionally, one can generate detailed pharmacological information, such as affinity measurements, down to almost single-molecule detection limits. The now accepted utilization of fluorescence-based readouts in high-throughput/high-content screening provides further evidence that fluorescent molecules offer a safer and more adaptable substitute to radioligands in molecular pharmacology and drug discovery. One such drug-target family that has received considerable attention are the GPCRs; this review therefore summarizes the most recent developments in the area of fluorescent ligand design for this important drug target. We assess recently reported fluorescent conjugates by adopting a receptor-family-based approach, highlighting some of the strengths and weaknesses of the individual molecules and their subsequent use. This review adds further strength to the arguments that fluorescent ligand design and synthesis requires careful planning and execution; providing examples illustrating that selection of the correct fluorescent dye, linker length/composition and geographic attachment point to the drug scaffold can all influence the ultimate selectivity and potency of the final conjugate when compared with its unlabelled precursor. When optimized appropriately, the resultant fluorescent conjugates have been successfully employed in an array of assay formats, including flow cytometry, fluorescence microscopy, FRET and scanning confocal microscopy. It is clear that fluorescently labelled GPCR ligands remain a developing and dynamic research arena.
Collapse
Affiliation(s)
- Andrea J Vernall
- School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | | | | |
Collapse
|
178
|
Hill SJ, May LT, Kellam B, Woolard J. Allosteric interactions at adenosine A(1) and A(3) receptors: new insights into the role of small molecules and receptor dimerization. Br J Pharmacol 2014; 171:1102-13. [PMID: 24024783 DOI: 10.1111/bph.12345] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/09/2013] [Accepted: 07/12/2013] [Indexed: 12/22/2022] Open
Abstract
The purine nucleoside adenosine is present in all cells in tightly regulated concentrations. It is released under a variety of physiological and pathophysiological conditions to facilitate protection and regeneration of tissues. Adenosine acts via specific GPCRs to either stimulate cyclic AMP formation, as exemplified by Gs -protein-coupled adenosine receptors (A2A and A2B ), or inhibit AC activity, in the case of Gi/o -coupled adenosine receptors (A1 and A3 ). Recent advances in our understanding of GPCR structure have provided insights into the conformational changes that occur during receptor activation following binding of agonists to orthosteric (i.e. at the same binding site as an endogenous modulator) and allosteric regulators to allosteric sites (i.e. at a site that is topographically distinct from the endogenous modulator). Binding of drugs to allosteric sites may lead to changes in affinity or efficacy, and affords considerable potential for increased selectivity in new drug development. Herein, we provide an overview of the properties of selective allosteric regulators of the adenosine A1 and A3 receptors, focusing on the impact of receptor dimerization, mechanistic approaches to single-cell ligand-binding kinetics and the effects of A1 - and A3 -receptor allosteric modulators on in vivo pharmacology.
Collapse
Affiliation(s)
- Stephen J Hill
- Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | | | | | | |
Collapse
|
179
|
Calebiro D, Maiellaro I. cAMP signaling microdomains and their observation by optical methods. Front Cell Neurosci 2014; 8:350. [PMID: 25389388 PMCID: PMC4211404 DOI: 10.3389/fncel.2014.00350] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/07/2014] [Indexed: 11/22/2022] Open
Abstract
The second messenger cyclic AMP (cAMP) is a major intracellular mediator of many hormones and neurotransmitters and regulates a myriad of cell functions, including synaptic plasticity in neurons. Whereas cAMP can freely diffuse in the cytosol, a growing body of evidence suggests the formation of cAMP gradients and microdomains near the sites of cAMP production, where cAMP signals remain apparently confined. The mechanisms responsible for the formation of such microdomains are subject of intensive investigation. The development of optical methods based on fluorescence resonance energy transfer (FRET), which allow a direct observation of cAMP signaling with high temporal and spatial resolution, is playing a fundamental role in elucidating the nature of such microdomains. Here, we will review the optical methods used for monitoring cAMP and protein kinase A (PKA) signaling in living cells, providing some examples of their application in neurons, and will discuss the major hypotheses on the formation of cAMP/PKA microdomains.
Collapse
Affiliation(s)
- Davide Calebiro
- Institute of Pharmacology and Toxicology, University of Würzburg Würzburg, Germany ; Bio-Imaging Center/Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg Würzburg, Germany
| | - Isabella Maiellaro
- Institute of Pharmacology and Toxicology, University of Würzburg Würzburg, Germany ; Bio-Imaging Center/Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg Würzburg, Germany
| |
Collapse
|
180
|
Role of 3D Structures in Understanding, Predicting, and Designing Molecular Interactions in the Chemokine Receptor Family. ACTA ACUST UNITED AC 2014. [DOI: 10.1007/7355_2014_77] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
181
|
González-Maeso J. Family a GPCR heteromers in animal models. Front Pharmacol 2014; 5:226. [PMID: 25346690 PMCID: PMC4191056 DOI: 10.3389/fphar.2014.00226] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 09/21/2014] [Indexed: 11/30/2022] Open
Affiliation(s)
- Javier González-Maeso
- Departments of Psychiatry and Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| |
Collapse
|
182
|
Baumgart F, Schütz GJ. Detecting protein association at the T cell plasma membrane. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:791-801. [PMID: 25300585 DOI: 10.1016/j.bbamcr.2014.09.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/18/2014] [Accepted: 09/29/2014] [Indexed: 10/24/2022]
Abstract
At the moment, many models on T cell signaling rely on results obtained via rather indirect methodologies, which makes direct comparison and conclusions to the in vivo situation difficult. Recently, a variety of new imaging methods were developed, which have the potential to directly shed light onto the mysteries of protein association at the T cell membrane. While the new modalities are extremely promising, for a broad readership it may be difficult to judge the results, since technological shortcomings are not always obvious. In this review article, we put key questions on the mechanism of protein interactions in the T cell plasma membrane into relation with techniques that allow to address such questions. We discuss applicability of the techniques, their strengths and weaknesses. This article is part of a Special Issue entitled: Nanoscale membrane organisation and signalling.
Collapse
Affiliation(s)
- Florian Baumgart
- Vienna University of Technology, Institute for Applied Physics, Wiedner Hauptstraße 8-10, 1040 Vienna, Austria
| | - Gerhard J Schütz
- Vienna University of Technology, Institute for Applied Physics, Wiedner Hauptstraße 8-10, 1040 Vienna, Austria.
| |
Collapse
|
183
|
Ciruela F, Jacobson KA, Fernández-Dueñas V. Portraying G protein-coupled receptors with fluorescent ligands. ACS Chem Biol 2014; 9:1918-28. [PMID: 25010291 PMCID: PMC4168789 DOI: 10.1021/cb5004042] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
The
thermodynamics of ligand–receptor interactions at the
surface of living cells represents a fundamental aspect of G protein-coupled
receptor (GPCR) biology; thus, its detailed elucidation constitutes
a challenge for modern pharmacology. Interestingly, fluorescent ligands
have been developed for a variety of GPCRs in order to monitor ligand–receptor
binding in living cells. Accordingly, new methodological strategies
derived from noninvasive fluorescence-based approaches, especially
fluorescence resonance energy transfer (FRET), have been successfully
developed to characterize ligand–receptor interactions. Importantly,
these technologies are supplanting more hazardous and expensive radioactive
binding assays. In addition, FRET-based tools have also become extremely
powerful approaches for visualizing receptor–receptor interactions
(i.e., GPCR oligomerization) in living cells. Thus, by means of the
synthesis of compatible fluorescent ligands these novel techniques
can be implemented to demonstrate the existence of GPCR oligomerization
not only in heterologous systems but also in native tissues. Finally,
there is no doubt that these methodologies would also be relevant
in drug discovery in order to develop new high-throughput screening
approaches or to identify new therapeutic targets. Overall, herein,
we provide a thorough assessment of all technical and biological aspects,
including strengths and weaknesses, of these fluorescence-based methodologies
when applied to the study of GPCR biology at the plasma membrane of
living cells.
Collapse
Affiliation(s)
- Francisco Ciruela
- Unitat
de Farmacologia, Departament Patologia i Terapèutica Experimental,
Facultat de Medicina, IDIBELL, Universitat de Barcelona, L’Hospitalet
de Llobregat, 08907 Barcelona, Spain
| | - Kenneth A. Jacobson
- Molecular
Recognition Section, Laboratory of Bioorganic Chemistry, National
Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Víctor Fernández-Dueñas
- Unitat
de Farmacologia, Departament Patologia i Terapèutica Experimental,
Facultat de Medicina, IDIBELL, Universitat de Barcelona, L’Hospitalet
de Llobregat, 08907 Barcelona, Spain
| |
Collapse
|
184
|
Huber T, Sakmar T. Chemical Biology Methods for Investigating G Protein-Coupled Receptor Signaling. ACTA ACUST UNITED AC 2014; 21:1224-37. [DOI: 10.1016/j.chembiol.2014.08.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/21/2014] [Accepted: 08/20/2014] [Indexed: 11/26/2022]
|
185
|
A new mechanism of allostery in a G protein-coupled receptor dimer. Nat Chem Biol 2014; 10:745-52. [PMID: 25108820 PMCID: PMC4138267 DOI: 10.1038/nchembio.1593] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 06/26/2014] [Indexed: 01/11/2023]
Abstract
SB269652 (1) is the first drug-like allosteric modulator of the
dopamine D2 receptor (D2R), but contains structural features
associated with orthosteric D2R antagonists. Using a functional complementation
system to control the identity of individual protomers within a dimeric D2R
complex, we converted the pharmacology of the interaction between SB269652 and dopamine
from allosteric to competitive by impairing ligand binding to one of the protomers,
indicating that the allostery requires D2R dimers. Additional experiments
identified a “bitopic” pose for SB269652 extending from the orthosteric
site into a secondary pocket at the extracellular end of the transmembrane (TM) domain,
involving TM2 and TM7. Engagement of this secondary pocket was a requirement for the
allosteric pharmacology of SB269652. This suggests a novel mechanism whereby a bitopic
ligand binds in an extended pose on one G protein-coupled receptor protomer to
allosterically modulate the binding of a ligand to the orthosteric site of a second
protomer.
Collapse
|
186
|
Tracking single molecules at work in living cells. Nat Chem Biol 2014; 10:524-32. [PMID: 24937070 DOI: 10.1038/nchembio.1558] [Citation(s) in RCA: 223] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 05/21/2014] [Indexed: 11/09/2022]
Abstract
Methods for imaging and tracking single molecules conjugated with fluorescent probes, called single-molecule tracking (SMT), are now providing researchers with the unprecedented ability to directly observe molecular behaviors and interactions in living cells. Current SMT methods are achieving almost the ultimate spatial precision and time resolution for tracking single molecules, determined by the currently available dyes. In cells, various molecular interactions and reactions occur as stochastic and probabilistic processes. SMT provides an ideal way to directly track these processes by observing individual molecules at work in living cells, leading to totally new views of the biochemical and molecular processes used by cells whether in signal transduction, gene regulation or formation and disintegration of macromolecular complexes. Here we review SMT methods, summarize the recent results obtained by SMT, including related superresolution microscopy data, and describe the special concerns when SMT applications are shifted from the in vitro paradigms to living cells.
Collapse
|
187
|
Sungkaworn T, Rieken F, Lohse MJ, Calebiro D. High-resolution spatiotemporal analysis of receptor dynamics by single-molecule fluorescence microscopy. J Vis Exp 2014:e51784. [PMID: 25145374 DOI: 10.3791/51784] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Single-molecule microscopy is emerging as a powerful approach to analyze the behavior of signaling molecules, in particular concerning those aspect (e.g., kinetics, coexistence of different states and populations, transient interactions), which are typically hidden in ensemble measurements, such as those obtained with standard biochemical or microscopy methods. Thus, dynamic events, such as receptor-receptor interactions, can be followed in real time in a living cell with high spatiotemporal resolution. This protocol describes a method based on labeling with small and bright organic fluorophores and total internal reflection fluorescence (TIRF) microscopy to directly visualize single receptors on the surface of living cells. This approach allows one to precisely localize receptors, measure the size of receptor complexes, and capture dynamic events such as transient receptor-receptor interactions. The protocol provides a detailed description of how to perform a single-molecule experiment, including sample preparation, image acquisition and image analysis. As an example, the application of this method to analyze two G-protein-coupled receptors, i.e., β2-adrenergic and γ-aminobutyric acid type B (GABAB) receptor, is reported. The protocol can be adapted to other membrane proteins and different cell models, transfection methods and labeling strategies.
Collapse
Affiliation(s)
- Titiwat Sungkaworn
- Institute of Pharmacology and Toxicology and Bio-Imaging Center/Rudolf Virchow Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Germany
| | - Finn Rieken
- Institute of Pharmacology and Toxicology and Bio-Imaging Center/Rudolf Virchow Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Germany
| | - Martin J Lohse
- Institute of Pharmacology and Toxicology and Bio-Imaging Center/Rudolf Virchow Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Germany
| | - Davide Calebiro
- Institute of Pharmacology and Toxicology and Bio-Imaging Center/Rudolf Virchow Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Germany;
| |
Collapse
|
188
|
Tian H, Naganathan S, Kazmi MA, Schwartz TW, Sakmar TP, Huber T. Bioorthogonal fluorescent labeling of functional G-protein-coupled receptors. Chembiochem 2014; 15:1820-9. [PMID: 25045132 DOI: 10.1002/cbic.201402193] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Indexed: 11/12/2022]
Abstract
Novel methods are required for site-specific, quantitative fluorescent labeling of G-protein-coupled receptors (GPCRs) and other difficult-to-express membrane proteins. Ideally, fluorescent probes should perturb the native structure and function as little as possible. We evaluated bioorthogonal reactions to label genetically encoded p-acetyl-L-phenylalanine (AcF) or p-azido-L-phenylalanine (azF) residues in receptors heterologously expressed in mammalian cells. We found that keto-selective reagents were not truly bioorthogonal, possibly owing to post-translational protein oxidation reactions. In contrast, the strain-promoted [3+2] azide-alkyne cycloaddition (SpAAC) with dibenzocyclooctyne (DIBO) reagents yielded stoichiometric conjugates with azF-rhodopsin while undergoing negligible background reactions. As one application of this technique, we used Alexa488-rhodopsin to measure the kinetics of ligand uptake and release in membrane-mimetic bicelles using a novel fluorescence-quenching assay.
Collapse
Affiliation(s)
- He Tian
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, 1230 York Avenue, New York, NY 10065 (USA)
| | | | | | | | | | | |
Collapse
|
189
|
Redka DS, Morizumi T, Elmslie G, Paranthaman P, Shivnaraine RV, Ellis J, Ernst OP, Wells JW. Coupling of g proteins to reconstituted monomers and tetramers of the M2 muscarinic receptor. J Biol Chem 2014; 289:24347-65. [PMID: 25023280 DOI: 10.1074/jbc.m114.559294] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
G protein-coupled receptors can be reconstituted as monomers in nanodiscs and as tetramers in liposomes. When reconstituted with G proteins, both forms enable an allosteric interaction between agonists and guanylyl nucleotides. Both forms, therefore, are candidates for the complex that controls signaling at the level of the receptor. To identify the biologically relevant form, reconstituted monomers and tetramers of the purified M2 muscarinic receptor were compared with muscarinic receptors in sarcolemmal membranes for the effect of guanosine 5'-[β,γ-imido]triphosphate (GMP-PNP) on the inhibition of N-[(3)H]methylscopolamine by the agonist oxotremorine-M. With monomers, a stepwise increase in the concentration of GMP-PNP effected a lateral, rightward shift in the semilogarithmic binding profile (i.e. a progressive decrease in the apparent affinity of oxotremorine-M). With tetramers and receptors in sarcolemmal membranes, GMP-PNP effected a vertical, upward shift (i.e. an apparent redistribution of sites from a state of high affinity to one of low affinity with no change in affinity per se). The data were analyzed in terms of a mechanistic scheme based on a ligand-regulated equilibrium between uncoupled and G protein-coupled receptors (the "ternary complex model"). The model predicts a rightward shift in the presence of GMP-PNP and could not account for the effects at tetramers in vesicles or receptors in sarcolemmal membranes. Monomers present a special case of the model in which agonists and guanylyl nucleotides interact within a complex that is both constitutive and stable. The results favor oligomers of the M2 receptor over monomers as the biologically relevant state for coupling to G proteins.
Collapse
Affiliation(s)
- Dar'ya S Redka
- From the Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Takefumi Morizumi
- the Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Gwendolynne Elmslie
- the Departments of Psychiatry and Pharmacology, Hershey Medical Center, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania 17033, and
| | - Pranavan Paranthaman
- From the Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Rabindra V Shivnaraine
- From the Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - John Ellis
- the Departments of Psychiatry and Pharmacology, Hershey Medical Center, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania 17033, and
| | - Oliver P Ernst
- the Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada, the Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - James W Wells
- From the Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada,
| |
Collapse
|
190
|
Ma Z, Du L, Li M. Toward fluorescent probes for G-protein-coupled receptors (GPCRs). J Med Chem 2014; 57:8187-203. [PMID: 24983484 DOI: 10.1021/jm401823z] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
G-protein-coupled receptors (GPCRs), a superfamily of cell-surface receptors that are the targets of about 40% of prescription drugs on the market, can sense numerous critical extracellular signals. Recent breakthroughs in structural biology, especially in holo-form X-ray crystal structures, have contributed to our understanding of GPCR signaling. However, actions of GPCRs at the cellular and molecular level, interactions between GPCRs, and the role of protein dynamics in receptor activities still remain controversial. To overcome these dilemmas, fluorescent probes of GPCRs have been employed, which have advantages of in vivo safety and real-time monitoring. Various probes that depend on specific mechanisms and/or technologies have been used to study GPCRs. The present review focuses on surveying the design and applications of fluorescent probes for GPCRs that are derived from small molecules or using protein-labeling techniques, as well as discussing some design strategies for new probes.
Collapse
Affiliation(s)
- Zhao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy, Shandong University , Jinan, Shandong 250012, China
| | | | | |
Collapse
|
191
|
Teichmann A, Gibert A, Lampe A, Grzesik P, Rutz C, Furkert J, Schmoranzer J, Krause G, Wiesner B, Schülein R. The specific monomer/dimer equilibrium of the corticotropin-releasing factor receptor type 1 is established in the endoplasmic reticulum. J Biol Chem 2014; 289:24250-62. [PMID: 24966326 DOI: 10.1074/jbc.m114.553644] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
G protein-coupled receptors (GPCRs) represent the most important drug targets. Although the smallest functional unit of a GPCR is a monomer, it became clear in the past decades that the vast majority of the receptors form dimers. Only very recently, however, data were presented that some receptors may in fact be expressed as a mixture of monomers and dimers and that the interaction of the receptor protomers is dynamic. To date, equilibrium measurements were restricted to the plasma membrane due to experimental limitations. We have addressed the question as to where this equilibrium is established for the corticotropin-releasing factor receptor type 1. By developing a novel approach to analyze single molecule fluorescence cross-correlation spectroscopy data for intracellular membrane compartments, we show that the corticotropin-releasing factor receptor type 1 has a specific monomer/dimer equilibrium that is already established in the endoplasmic reticulum (ER). It remains constant at the plasma membrane even following receptor activation. Moreover, we demonstrate for seven additional GPCRs that they are expressed in specific but substantially different monomer/dimer ratios. Although it is well known that proteins may dimerize in the ER in principle, our data show that the ER is also able to establish the specific monomer/dimer ratios of GPCRs, which sheds new light on the functions of this compartment.
Collapse
Affiliation(s)
- Anke Teichmann
- From the Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Arthur Gibert
- From the Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - André Lampe
- From the Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Paul Grzesik
- From the Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Claudia Rutz
- From the Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Jens Furkert
- From the Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Jan Schmoranzer
- From the Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Gerd Krause
- From the Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Burkhard Wiesner
- From the Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Ralf Schülein
- From the Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| |
Collapse
|
192
|
Stephens B, Handel TM. Chemokine receptor oligomerization and allostery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 115:375-420. [PMID: 23415099 DOI: 10.1016/b978-0-12-394587-7.00009-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oligomerization of chemokine receptors has been reported to influence many aspects of receptor function through allosteric communication between receptor protomers. Allosteric interactions within chemokine receptor hetero-oligomers have been shown to cause negative cooperativity in the binding of chemokines and to inhibit receptor activation in the case of some receptor pairs. Other receptor pairs can cause enhanced signaling and even activate entirely new, hetero-oligomer-specific signaling complexes and responses downstream of receptor activation. Many mechanisms contribute to these effects including direct allosteric coupling between the receptors, G protein-mediated allostery, G protein stealing, ligand sequestration, and recruitment of new intracellular proteins by exposing unique binding interfaces on the oligomerized receptors. These effects present both challenges as well as exciting opportunities for drug discovery. One of the most difficult challenges will involve determining if and when hetero-oligomers versus homomeric receptors are involved in specific disease states.
Collapse
Affiliation(s)
- Bryan Stephens
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, California, USA
| | | |
Collapse
|
193
|
Jäntti MH, Mandrika I, Kukkonen JP. Human orexin/hypocretin receptors form constitutive homo- and heteromeric complexes with each other and with human CB1 cannabinoid receptors. Biochem Biophys Res Commun 2014; 445:486-90. [PMID: 24530395 DOI: 10.1016/j.bbrc.2014.02.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 02/06/2014] [Indexed: 12/17/2022]
Abstract
Human OX1 orexin receptors have been shown to homodimerize and they have also been suggested to heterodimerize with CB1 cannabinoid receptors. The latter has been suggested to be important for orexin receptor responses and trafficking. In this study, we wanted to assess the ability of the other combinations of receptors to also form similar complexes. Vectors for expression of human OX1, OX2 and CB1 receptors, C-terminally fused with either Renilla luciferase or GFP(2) green fluorescent protein variant, were generated. The constructs were transiently expressed in Chinese hamster ovary cells, and constitutive dimerization between the receptors was assessed by bioluminescence energy transfer (BRET). Orexin receptor subtypes readily formed homo- and hetero(di)mers, as suggested by significant BRET signals. CB1 receptors formed homodimers, and they also heterodimerized with both orexin receptors. Interestingly, BRET efficiency was higher for homodimers than for almost all heterodimers. This is likely to be due to the geometry of the interaction; the putatively symmetric dimers may place the C-termini in a more suitable orientation in homomers. Fusion of luciferase to an orexin receptor and GFP(2) to CB1 produced more effective BRET than the opposite fusions, also suggesting differences in geometry. Similar was seen for the OX1-OX2 interaction. In conclusion, orexin receptors have a significant propensity to make homo- and heterodi-/oligomeric complexes. However, it is unclear whether this affects their signaling. As orexin receptors efficiently signal via endocannabinoid production to CB1 receptors, dimerization could be an effective way of forming signal complexes with optimal cannabinoid concentrations available for cannabinoid receptors.
Collapse
Affiliation(s)
- Maria H Jäntti
- Department of Veterinary Biosciences, POB 66, FIN-00014 University of Helsinki, Finland.
| | - Ilona Mandrika
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, Riga LV 1067, Latvia.
| | - Jyrki P Kukkonen
- Department of Veterinary Biosciences, POB 66, FIN-00014 University of Helsinki, Finland.
| |
Collapse
|
194
|
Ferré S, Casadó V, Devi LA, Filizola M, Jockers R, Lohse MJ, Milligan G, Pin JP, Guitart X. G protein-coupled receptor oligomerization revisited: functional and pharmacological perspectives. Pharmacol Rev 2014; 66:413-34. [PMID: 24515647 PMCID: PMC3973609 DOI: 10.1124/pr.113.008052] [Citation(s) in RCA: 442] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Most evidence indicates that, as for family C G protein-coupled receptors (GPCRs), family A GPCRs form homo- and heteromers. Homodimers seem to be a predominant species, with potential dynamic formation of higher-order oligomers, particularly tetramers. Although monomeric GPCRs can activate G proteins, the pentameric structure constituted by one GPCR homodimer and one heterotrimeric G protein may provide a main functional unit, and oligomeric entities can be viewed as multiples of dimers. It still needs to be resolved if GPCR heteromers are preferentially heterodimers or if they are mostly constituted by heteromers of homodimers. Allosteric mechanisms determine a multiplicity of possible unique pharmacological properties of GPCR homomers and heteromers. Some general mechanisms seem to apply, particularly at the level of ligand-binding properties. In the frame of the dimer-cooperativity model, the two-state dimer model provides the most practical method to analyze ligand-GPCR interactions when considering receptor homomers. In addition to ligand-binding properties, unique properties for each GPCR oligomer emerge in relation to different intrinsic efficacy of ligands for different signaling pathways (functional selectivity). This gives a rationale for the use of GPCR oligomers, and particularly heteromers, as novel targets for drug development. Herein, we review the functional and pharmacological properties of GPCR oligomers and provide some guidelines for the application of discrete direct screening and high-throughput screening approaches to the discovery of receptor-heteromer selective compounds.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Department of Health and Human Services, 333 Cassell Drive, Baltimore, Maryland 21224.
| | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Labilloy A, Youker RT, Bruns JR, Kukic I, Kiselyov K, Halfter W, Finegold D, do Monte SJH, Weisz OA. Altered dynamics of a lipid raft associated protein in a kidney model of Fabry disease. Mol Genet Metab 2014; 111:184-92. [PMID: 24215843 PMCID: PMC3946758 DOI: 10.1016/j.ymgme.2013.10.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 10/12/2013] [Indexed: 02/07/2023]
Abstract
Accumulation of globotriaosylceramide (Gb3) and other neutral glycosphingolipids with galactosyl residues is the hallmark of Fabry disease, a lysosomal storage disorder caused by deficiency of the enzyme alpha-galactosidase A (α-gal A). These lipids are incorporated into the plasma membrane and intracellular membranes, with a preference for lipid rafts. Disruption of raft mediated cell processes is implicated in the pathogenesis of several human diseases, but little is known about the effects of the accumulation of glycosphingolipids on raft dynamics in the context of Fabry disease. Using siRNA technology, we have generated a polarized renal epithelial cell model of Fabry disease in Madin-Darby canine kidney cells. These cells present increased levels of Gb3 and enlarged lysosomes, and progressively accumulate zebra bodies. The polarized delivery of both raft-associated and raft-independent proteins was unaffected by α-gal A knockdown, suggesting that accumulation of Gb3 does not disrupt biosynthetic trafficking pathways. To assess the effect of α-gal A silencing on lipid raft dynamics, we employed number and brightness (N&B) analysis to measure the oligomeric status and mobility of the model glycosylphosphatidylinositol (GPI)-anchored protein GFP-GPI. We observed a significant increase in the oligomeric size of antibody-induced clusters of GFP-GPI at the plasma membrane of α-gal A silenced cells compared with control cells. Our results suggest that the interaction of GFP-GPI with lipid rafts may be altered in the presence of accumulated Gb3. The implications of our results with respect to the pathogenesis of Fabry disease are discussed.
Collapse
Affiliation(s)
- Anatália Labilloy
- Renal-Electrolyte Division, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Ciência sem Fronteiras, CNPq, Brazil
| | - Robert T Youker
- Renal-Electrolyte Division, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jennifer R Bruns
- Renal-Electrolyte Division, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ira Kukic
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Willi Halfter
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - David Finegold
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | - Ora A Weisz
- Renal-Electrolyte Division, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
196
|
Jiang X, Dias JA, He X. Structural biology of glycoprotein hormones and their receptors: insights to signaling. Mol Cell Endocrinol 2014; 382:424-451. [PMID: 24001578 DOI: 10.1016/j.mce.2013.08.021] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/20/2013] [Accepted: 08/24/2013] [Indexed: 01/18/2023]
Abstract
This article reviews the progress made in the field of glycoprotein hormones (GPH) and their receptors (GPHR) by several groups of structural biologists including ourselves aiming to gain insight into GPH signaling mechanisms. The GPH family consists of four members, with follicle-stimulating hormone (FSH) being the prototypic member. GPH members belong to the cystine-knot growth factor superfamily, and their receptors (GPHR), possessing unusually large N-terminal ectodomains, belong to the G-protein coupled receptor Family A. GPHR ectodomains can be divided into two subdomains: a high-affinity hormone binding subdomain primarily centered on the N-terminus, and a second subdomain that is located on the C-terminal region of the ectodomain that is involved in signal specificity. The two subdomains unexpectedly form an integral structure comprised of leucine-rich repeats (LRRs). Following the structure determination of hCG in 1994, the field of FSH structural biology has progressively advanced. Initially, the FSH structure was determined in partially glycosylated free form in 2001, followed by a structure of FSH bound to a truncated FSHR ectodomain in 2005, and the structure of FSH bound to the entire ectodomain in 2012. Comparisons of the structures in three forms led a proposal of a two-step monomeric receptor activation mechanism. First, binding of FSH to the FSHR high-affinity hormone-binding subdomain induces a conformational change in the hormone to form a binding pocket that is specific for a sulfated-tyrosine found as sTyr 335 in FSHR. Subsequently, the sTyr is drawn into the newly formed binding pocket, producing a lever effect on a helical pivot whereby the docking sTyr provides as the 'pull & lift' force. The pivot helix is flanked by rigid LRRs and locked by two disulfide bonds on both sides: the hormone-binding subdomain on one side and the last short loop before the first transmembrane helix on the other side. The lift of the sTyr loop frees the tethered extracellular loops of the 7TM domain, thereby releasing a putative inhibitory influence of the ectodomain, ultimately leading to the activating conformation of the 7TM domain. Moreover, the data lead us to propose that FSHR exists as a trimer and to present an FSHR activation mechanism consistent with the observed trimeric crystal form. A trimeric receptor provides resolution of the enigmatic, but important, biological roles played by GPH residues that are removed from the primary FSH-binding site, as well as several important GPCR phenomena, including negative cooperativity and asymmetric activation. Further reflection pursuant to this review process revealed additional novel structural characteristics such as the identification of a 'seat' sequence in GPH. Together with the 'seatbelt', the 'seat' enables a common heteodimeric mode of association of the common α subunit non-covalently and non-specifically with each of the three different β subunits. Moreover, it was possible to establish a dimensional order that can be used to estimate LRR curvatures. A potential binding pocket for small molecular allosteric modulators in the FSHR 7TM domain has also been identified.
Collapse
Affiliation(s)
- Xuliang Jiang
- EMD Serono Research & Development Institute, Billerica, MA 01821, United States.
| | - James A Dias
- Department of Biomedical Sciences, School of Public Health, University at Albany-SUNY, Albany, NY 12222, United States
| | - Xiaolin He
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| |
Collapse
|
197
|
Johnston JM, Filizola M. Beyond standard molecular dynamics: investigating the molecular mechanisms of G protein-coupled receptors with enhanced molecular dynamics methods. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 796:95-125. [PMID: 24158803 PMCID: PMC4074508 DOI: 10.1007/978-94-007-7423-0_6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The majority of biological processes mediated by G Protein-Coupled Receptors (GPCRs) take place on timescales that are not conveniently accessible to standard molecular dynamics (MD) approaches, notwithstanding the current availability of specialized parallel computer architectures, and efficient simulation algorithms. Enhanced MD-based methods have started to assume an important role in the study of the rugged energy landscape of GPCRs by providing mechanistic details of complex receptor processes such as ligand recognition, activation, and oligomerization. We provide here an overview of these methods in their most recent application to the field.
Collapse
Affiliation(s)
- Jennifer M. Johnston
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Marta Filizola
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
198
|
Fuxe K, Borroto-Escuela DO, Romero-Fernandez W, Palkovits M, Tarakanov AO, Ciruela F, Agnati LF. Moonlighting proteins and protein-protein interactions as neurotherapeutic targets in the G protein-coupled receptor field. Neuropsychopharmacology 2014; 39:131-55. [PMID: 24105074 PMCID: PMC3857668 DOI: 10.1038/npp.2013.242] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 07/04/2013] [Accepted: 07/05/2013] [Indexed: 12/28/2022]
Abstract
There is serious interest in understanding the dynamics of the receptor-receptor and receptor-protein interactions in space and time and their integration in GPCR heteroreceptor complexes of the CNS. Moonlighting proteins are special multifunctional proteins because they perform multiple autonomous, often unrelated, functions without partitioning into different protein domains. Moonlighting through receptor oligomerization can be operationally defined as an allosteric receptor-receptor interaction, which leads to novel functions of at least one receptor protomer. GPCR-mediated signaling is a more complicated process than previously described as every GPCR and GPCR heteroreceptor complex requires a set of G protein interacting proteins, which interacts with the receptor in an orchestrated spatio-temporal fashion. GPCR heteroreceptor complexes with allosteric receptor-receptor interactions operating through the receptor interface have become major integrative centers at the molecular level and their receptor protomers act as moonlighting proteins. The GPCR heteroreceptor complexes in the CNS have become exciting new targets for neurotherapeutics in Parkinson's disease, schizophrenia, drug addiction, and anxiety and depression opening a new field in neuropsychopharmacology.
Collapse
Affiliation(s)
- Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet,, Stockholm, Sweden
| | | | | | - Miklós Palkovits
- Department of Anatomy, Histology and Embryology, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Alexander O Tarakanov
- Russian Academy of Sciences, St. Petersburg Institute for Informatics and Automation, Saint Petersburg, Russia
| | - Francisco Ciruela
- Facultat de Medicina, Departament de Patologia i Terapèutica Experimental IDIBELL-Universitat de Barcelona, L'Hospitalet de Llobregat, Unitat de Farmacologia, Barcelona, Spain
| | | |
Collapse
|
199
|
Sridharan R, Zuber J, Connelly SM, Mathew E, Dumont ME. Fluorescent approaches for understanding interactions of ligands with G protein coupled receptors. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1838:15-33. [PMID: 24055822 PMCID: PMC3926105 DOI: 10.1016/j.bbamem.2013.09.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 09/03/2013] [Accepted: 09/08/2013] [Indexed: 11/18/2022]
Abstract
G protein coupled receptors are responsible for a wide variety of signaling responses in diverse cell types. Despite major advances in the determination of structures of this class of receptors, the underlying mechanisms by which binding of different types of ligands specifically elicits particular signaling responses remain unclear. The use of fluorescence spectroscopy can provide important information about the process of ligand binding and ligand dependent conformational changes in receptors, especially kinetic aspects of these processes that can be difficult to extract from X-ray structures. We present an overview of the extensive array of fluorescent ligands that have been used in studies of G protein coupled receptors and describe spectroscopic approaches for assaying binding and probing the environment of receptor-bound ligands with particular attention to examples involving yeast pheromone receptors. In addition, we discuss the use of fluorescence spectroscopy for detecting and characterizing conformational changes in receptors induced by the binding of ligands. Such studies have provided strong evidence for diversity of receptor conformations elicited by different ligands, consistent with the idea that GPCRs are not simple on and off switches. This diversity of states constitutes an underlying mechanistic basis for biased agonism, the observation that different stimuli can produce different responses from a single receptor. It is likely that continued technical advances will allow fluorescence spectroscopy to play an important role in continued probing of structural transitions in G protein coupled receptors. This article is part of a Special Issue entitled: Structural and biophysical characterisation of membrane protein-ligand binding.
Collapse
Affiliation(s)
- Rajashri Sridharan
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Jeffrey Zuber
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Sara M. Connelly
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Elizabeth Mathew
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Mark E. Dumont
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pediatrics, P.O. Box 777, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
200
|
Mondal S, Khelashvili G, Johner N, Weinstein H. How the dynamic properties and functional mechanisms of GPCRs are modulated by their coupling to the membrane environment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 796:55-74. [PMID: 24158801 DOI: 10.1007/978-94-007-7423-0_4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Experimental observations of the dependence of function and organization of G protein-coupled receptors (GPCRs) on their lipid environment have stimulated new quantitative studies of the coupling between the proteins and the membrane. It is important to develop such a quantitative understanding at the molecular level because the effects of the coupling are seen to be physiologically and clinically significant. Here we review findings that offer insight into how membrane-GPCR coupling is connected to the structural characteristics of the GPCR, from sequence to 3D structural detail, and how this coupling is involved in the actions of ligands on the receptor. The application of a recently developed computational approach designed for quantitative evaluation of membrane remodeling and the energetics of membrane-protein interactions brings to light the importance of the radial asymmetry of the membrane-facing surface of GPCRs in their interaction with the surrounding membrane. As the radial asymmetry creates adjacencies of hydrophobic and polar residues at specific sites of the GPCR, the ability of membrane remodeling to achieve complete hydrophobic matching is limited, and the residual mismatch carries a significant energy cost. The adjacencies are shown to be affected by ligand-induced conformational changes. Thus, functionally important organization of GPCRs in the cell membrane can depend both on ligand-determined properties and on the lipid composition of various membrane regions with different remodeling capacities. That this functionally important reorganization can be driven by oligomerization patterns that reduce the energy cost of the residual mismatch, suggests a new perspective on GPCR dimerization and ligand-GPCR interactions. The relation between the modulatory effects on GPCRs from the binding of specific cell-membrane components, e.g., cholesterol, and those produced by the non-local energetics of hydrophobic mismatch are discussed in this context.
Collapse
Affiliation(s)
- Sayan Mondal
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, Room E-509, 1300 York Avenue, 10065, New York City, NY, USA
| | | | | | | |
Collapse
|