51
|
Cullen PJ, Steinberg F. To degrade or not to degrade: mechanisms and significance of endocytic recycling. Nat Rev Mol Cell Biol 2018; 19:679-696. [PMID: 30194414 DOI: 10.1038/s41580-018-0053-7] [Citation(s) in RCA: 383] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Newly endocytosed integral cell surface proteins are typically either directed for degradation or subjected to recycling back to the plasma membrane. The sorting of integral cell surface proteins, including signalling receptors, nutrient transporters, ion channels, adhesion molecules and polarity markers, within the endolysosomal network for recycling is increasingly recognized as an essential feature in regulating the complexities of physiology at the cell, tissue and organism levels. Historically, endocytic recycling has been regarded as a relatively passive process, where the majority of internalized integral proteins are recycled via a nonspecific sequence-independent 'bulk membrane flow' pathway. Recent work has increasingly challenged this view. The discovery of sequence-specific sorting motifs and the identification of cargo adaptors and associated coat complexes have begun to uncover the highly orchestrated nature of endosomal cargo recycling, thereby providing new insight into the function and (patho)physiology of this process.
Collapse
Affiliation(s)
- Peter J Cullen
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, UK.
| | - Florian Steinberg
- Center for Biological Systems Analysis, Albert Ludwigs Universitaet Freiburg, Freiburg im Breisgau, Germany.
| |
Collapse
|
52
|
Luttrell LM, Wang J, Plouffe B, Smith JS, Yamani L, Kaur S, Jean-Charles PY, Gauthier C, Lee MH, Pani B, Kim J, Ahn S, Rajagopal S, Reiter E, Bouvier M, Shenoy SK, Laporte SA, Rockman HA, Lefkowitz RJ. Manifold roles of β-arrestins in GPCR signaling elucidated with siRNA and CRISPR/Cas9. Sci Signal 2018; 11:11/549/eaat7650. [PMID: 30254056 DOI: 10.1126/scisignal.aat7650] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) use diverse mechanisms to regulate the mitogen-activated protein kinases ERK1/2. β-Arrestins (βArr1/2) are ubiquitous inhibitors of G protein signaling, promoting GPCR desensitization and internalization and serving as scaffolds for ERK1/2 activation. Studies using CRISPR/Cas9 to delete βArr1/2 and G proteins have cast doubt on the role of β-arrestins in activating specific pools of ERK1/2. We compared the effects of siRNA-mediated knockdown of βArr1/2 and reconstitution with βArr1/2 in three different parental and CRISPR-derived βArr1/2 knockout HEK293 cell pairs to assess the effect of βArr1/2 deletion on ERK1/2 activation by four Gs-coupled GPCRs. In all parental lines with all receptors, ERK1/2 stimulation was reduced by siRNAs specific for βArr2 or βArr1/2. In contrast, variable effects were observed with CRISPR-derived cell lines both between different lines and with activation of different receptors. For β2 adrenergic receptors (β2ARs) and β1ARs, βArr1/2 deletion increased, decreased, or had no effect on isoproterenol-stimulated ERK1/2 activation in different CRISPR clones. ERK1/2 activation by the vasopressin V2 and follicle-stimulating hormone receptors was reduced in these cells but was enhanced by reconstitution with βArr1/2. Loss of desensitization and receptor internalization in CRISPR βArr1/2 knockout cells caused β2AR-mediated stimulation of ERK1/2 to become more dependent on G proteins, which was reversed by reintroducing βArr1/2. These data suggest that βArr1/2 function as a regulatory hub, determining the balance between mechanistically different pathways that result in activation of ERK1/2, and caution against extrapolating results obtained from βArr1/2- or G protein-deleted cells to GPCR behavior in native systems.
Collapse
Affiliation(s)
- Louis M Luttrell
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA.,Research Service of the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Jialu Wang
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Bianca Plouffe
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C IJ4, Canada
| | - Jeffrey S Smith
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Lama Yamani
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Suneet Kaur
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Christophe Gauthier
- Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, CNRS, Université de Tours, 37380 Nouzilly, France
| | - Mi-Hye Lee
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Biswaranjan Pani
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Jihee Kim
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Seungkirl Ahn
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Sudarshan Rajagopal
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Eric Reiter
- Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, CNRS, Université de Tours, 37380 Nouzilly, France
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C IJ4, Canada
| | - Sudha K Shenoy
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Stéphane A Laporte
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Robert J Lefkowitz
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA. .,Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA.,Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
53
|
Agwuegbo UC, Jonas KC. Molecular and functional insights into gonadotropin hormone receptor dimerization and oligomerization. ACTA ACUST UNITED AC 2018; 70:539-548. [PMID: 30226027 DOI: 10.23736/s0026-4784.18.04287-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The gonadotropin hormones, follicle stimulating hormone and luteinizing hormone, are essential for reproduction. They work in concert to control multiple aspects of gonadal function to ultimately produce meiotically competent and fertilizable gametes, provide the optimal endometrial environment and support for implantation and maintain pregnancy via progesterone production throughout the first trimester of pregnancy. These complex and multidimensional functions are mediated via the gonadotropin hormone receptors, luteinizing hormone receptor and follicle stimulating hormone receptor, Class A G protein-coupled receptors (GPCR), which couple to multiple G protein-dependent and independent signal pathways to control these physiological processes. Over the last two decades, a plethora of experimental evidence has shown that GPCRs can associate to form dimers and oligomers. This association provides a means of mediating the diverse functional requirements of a single receptor subtype and for the gonadotropin hormone receptors, has been shown to alter the pharmacology and signal activation profile of these receptors. This review will detail the historical and current evidence detailing the formation of gonadotropin hormone receptor homomers and heteromers. We will discuss the functional insights gained from in vitro and in vivo studies, and the potential impact in modulating reproductive health and disease.
Collapse
Affiliation(s)
- Uche C Agwuegbo
- Vascular Biology Research Center, St George's University of London, London, UK
| | - Kim C Jonas
- Vascular Biology Research Center, St George's University of London, London, UK - .,Institute of Medical and Biomedical Education, St George's University of London, London, UK
| |
Collapse
|
54
|
Ulloa-Aguirre A, Reiter E, Crépieux P. FSH Receptor Signaling: Complexity of Interactions and Signal Diversity. Endocrinology 2018; 159:3020-3035. [PMID: 29982321 DOI: 10.1210/en.2018-00452] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022]
Abstract
FSH is synthesized in the pituitary by gonadotrope cells. By binding to and interacting with its cognate receptor [FSH receptor (FSHR)] in the gonads, this gonadotropin plays a key role in the control of gonadal function and reproduction. Upon activation, the FSHR undergoes conformational changes leading to transduction of intracellular signals, including dissociation of G protein complexes into components and activation of several associated interacting partners, which concertedly regulate downstream effectors. The canonical Gs/cAMP/protein kinase A pathway, considered for a long time as the sole effector of FSHR-mediated signaling, is now viewed as one of several mechanisms employed by this receptor to transduce intracellular signals in response to the FSH stimulus. This complex network of signaling pathways allows for a fine-tuning regulation of the gonadotropic stimulus, where activation/inhibition of its multiple components vary depending on the cell context, cell developmental stage, and concentration of associated receptors and corresponding ligands. Activation of these multiple signaling modules eventually converge to the hormone-integrated biological response, including survival, proliferation and differentiation of target cells, synthesis and secretion of paracrine/autocrine regulators, and, at the molecular level, functional selectivity and differential gene expression. In this mini-review, we discuss the complexity of FSHR-mediated intracellular signals activated in response to ligand stimulation. A better understanding of the signaling pathways involved in FSH action might potentially influence the development of new therapeutic strategies for reproductive disorders.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Eric Reiter
- Biology and Bioinformatics of Signaling Systems Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Nouzilly, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Nouzilly, France
- Université François Rabelais, Nouzilly, France
| | - Pascale Crépieux
- Biology and Bioinformatics of Signaling Systems Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Nouzilly, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Nouzilly, France
- Université François Rabelais, Nouzilly, France
| |
Collapse
|
55
|
Nooh MM, Mancarella S, Bahouth SW. Novel Paradigms Governing β1-Adrenergic Receptor Trafficking in Primary Adult Rat Cardiac Myocytes. Mol Pharmacol 2018; 94:862-875. [PMID: 29848777 PMCID: PMC6022806 DOI: 10.1124/mol.118.112045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/23/2018] [Indexed: 12/11/2022] Open
Abstract
The β1-adrenergic receptor (β1-AR) is a major cardiac G protein-coupled receptor, which mediates cardiac actions of catecholamines and is involved in genesis and treatment of numerous cardiovascular disorders. In mammalian cells, catecholamines induce the internalization of the β1-AR into endosomes and their removal promotes the recycling of the endosomal β1-AR back to the plasma membrane; however, whether these redistributive processes occur in terminally differentiated cells is unknown. Compartmentalization of the β1-AR in response to β-agonists and antagonists was determined by confocal microscopy in primary adult rat ventricular myocytes (ARVMs), which are terminally differentiated myocytes with unique structures such as transverse tubules (T-tubules) and contractile sarcomeres. In unstimulated ARVMs, the fluorescently labeled β1-AR was expressed on the external membrane (the sarcolemma) of cardiomyocytes. Exposing ARVMs to isoproterenol redistributed surface β1-ARs into small (∼225-250 nm) regularly spaced internal punctate structures that overlapped with puncta stained by Di-8 ANEPPS, a membrane-impermeant T-tubule-specific dye. Replacing the β-agonist with the β-blocker alprenolol, induced the translocation of the wild-type β1-AR from these punctate structures back to the plasma membrane. This step was dependent on two barcodes, namely, the type-1 PDZ binding motif and serine at position 312 of the β1-AR, which is phosphorylated by a pool of cAMP-dependent protein kinases anchored at the type-1 PDZ of the β1-AR. These data show that redistribution of the β1-AR in ARVMs from internal structures back to the plasma membrane was mediated by a novel sorting mechanism, which might explain unique aspects of cardiac β1-AR signaling under normal or pathologic conditions.
Collapse
Affiliation(s)
- Mohammed M Nooh
- Departments of Pharmacology (M.M.N., S.W.B.) and Physiology (S.M.), The University of Tennessee Health Sciences Center, Memphis, Tennessee; and Department of Biochemistry, Faculty of Pharmacy Cairo University, Cairo, Egypt (M.M.N.)
| | - Salvatore Mancarella
- Departments of Pharmacology (M.M.N., S.W.B.) and Physiology (S.M.), The University of Tennessee Health Sciences Center, Memphis, Tennessee; and Department of Biochemistry, Faculty of Pharmacy Cairo University, Cairo, Egypt (M.M.N.)
| | - Suleiman W Bahouth
- Departments of Pharmacology (M.M.N., S.W.B.) and Physiology (S.M.), The University of Tennessee Health Sciences Center, Memphis, Tennessee; and Department of Biochemistry, Faculty of Pharmacy Cairo University, Cairo, Egypt (M.M.N.)
| |
Collapse
|
56
|
Integration of GPCR Signaling and Sorting from Very Early Endosomes via Opposing APPL1 Mechanisms. Cell Rep 2018; 21:2855-2867. [PMID: 29212031 PMCID: PMC5732320 DOI: 10.1016/j.celrep.2017.11.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/18/2017] [Accepted: 11/03/2017] [Indexed: 01/10/2023] Open
Abstract
Endocytic trafficking is a critical mechanism for cells to decode complex signaling pathways, including those activated by G-protein-coupled receptors (GPCRs). Heterogeneity in the endosomal network enables GPCR activity to be spatially restricted between early endosomes (EEs) and the recently discovered endosomal compartment, the very early endosome (VEE). However, the molecular machinery driving GPCR activity from the VEE is unknown. Using luteinizing hormone receptor (LHR) as a prototype GPCR for this compartment, along with additional VEE-localized GPCRs, we identify a role for the adaptor protein APPL1 in rapid recycling and endosomal cAMP signaling without impacting the EE-localized β2-adrenergic receptor. LHR recycling is driven by receptor-mediated Gαs/cAMP signaling from the VEE and PKA-dependent phosphorylation of APPL1 at serine 410. Receptor/Gαs endosomal signaling is localized to microdomains of heterogeneous VEE populations and regulated by APPL1 phosphorylation. Our study uncovers a highly integrated inter-endosomal communication system enabling cells to tightly regulate spatially encoded signaling. GPCRs that internalize to very early endosomes (VEEs) require APPL1 to recycle Receptor recycling is driven by cAMP/PKA to phosphorylate serine 410 on APPL1 cAMP signaling from GPCRs, such as LHR, occurs from distinct VEE microdomains APPL1 limits VEE cAMP signaling via opposing mechanisms required for GPCR sorting
Collapse
|
57
|
Gorvin CM. Insights into calcium-sensing receptor trafficking and biased signalling by studies of calcium homeostasis. J Mol Endocrinol 2018; 61:R1-R12. [PMID: 29599414 DOI: 10.1530/jme-18-0049] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/29/2018] [Indexed: 12/18/2022]
Abstract
The calcium-sensing receptor (CASR) is a class C G-protein-coupled receptor (GPCR) that detects extracellular calcium concentrations, and modulates parathyroid hormone secretion and urinary calcium excretion to maintain calcium homeostasis. The CASR utilises multiple heterotrimeric G-proteins to mediate signalling effects including activation of intracellular calcium release; mitogen-activated protein kinase (MAPK) pathways; membrane ruffling; and inhibition of cAMP production. By studying germline mutations in the CASR and proteins within its signalling pathway that cause hyper- and hypocalcaemic disorders, novel mechanisms governing GPCR signalling and trafficking have been elucidated. This review focusses on two recently described pathways that provide novel insights into CASR signalling and trafficking mechanisms. The first, identified by studying a CASR gain-of-function mutation that causes autosomal dominant hypocalcaemia (ADH), demonstrated a structural motif located between the third transmembrane domain and the second extracellular loop of the CASR that mediates biased signalling by activating a novel β-arrestin-mediated G-protein-independent pathway. The second, in which the mechanism by which adaptor protein-2 σ-subunit (AP2σ) mutations cause familial hypocalciuric hypercalcaemia (FHH) was investigated, demonstrated that AP2σ mutations impair CASR internalisation and reduce multiple CASR-mediated signalling pathways. Furthermore, these studies showed that the CASR can signal from the cell surface using multiple G-protein pathways, whilst sustained signalling is mediated only by the Gq/11 pathway. Thus, studies of FHH- and ADH-associated mutations have revealed novel steps by which CASR mediates signalling and compartmental bias, and these pathways could provide new targets for therapies for patients with calcaemic disorders.
Collapse
Affiliation(s)
- Caroline M Gorvin
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
| |
Collapse
|
58
|
Hanyaloglu AC. Advances in Membrane Trafficking and Endosomal Signaling of G Protein-Coupled Receptors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 339:93-131. [PMID: 29776606 DOI: 10.1016/bs.ircmb.2018.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The integration of GPCR signaling with membrane trafficking, as a single orchestrated system, is a theme increasingly evident with the growing reports of GPCR endosomal signaling. Once viewed as a mechanism to regulate cell surface heterotrimeric G protein signaling, the endocytic trafficking system is complex, highly compartmentalized, yet deeply interconnected with cell signaling. The organization of receptors into distinct plasma membrane signalosomes, biochemically distinct endosomal populations, endosomal microdomains, and its communication with distinct subcellular organelles such as the Golgi provides multiple unique signaling platforms that are critical for specifying receptor function physiologically and pathophysiologically. In this chapter I discuss our emerging understanding in the endocytic trafficking systems employed by GPCRs and their novel roles in spatial control of signaling. Given the extensive roles that GPCRs play in vivo, these evolving models are starting to provide mechanistic understanding of distinct diseases and provide novel therapeutic avenues that are proving to be viable targets.
Collapse
Affiliation(s)
- Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, London, United Kingdom.
| |
Collapse
|
59
|
Buenaventura T, Kanda N, Douzenis PC, Jones B, Bloom SR, Chabosseau P, Corrêa IR, Bosco D, Piemonti L, Marchetti P, Johnson PR, Shapiro AMJ, Rutter GA, Tomas A. A Targeted RNAi Screen Identifies Endocytic Trafficking Factors That Control GLP-1 Receptor Signaling in Pancreatic β-Cells. Diabetes 2018; 67:385-399. [PMID: 29284659 DOI: 10.2337/db17-0639] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 12/19/2017] [Indexed: 11/13/2022]
Abstract
The glucagon-like peptide 1 (GLP-1) receptor (GLP-1R) is a key target for type 2 diabetes (T2D) treatment. Because endocytic trafficking of agonist-bound receptors is one of the most important routes for regulation of receptor signaling, a better understanding of this process may facilitate the development of new T2D therapeutic strategies. Here, we screened 29 proteins with known functions in G protein-coupled receptor trafficking for their role in GLP-1R potentiation of insulin secretion in pancreatic β-cells. We identify five (clathrin, dynamin1, AP2, sorting nexins [SNX] SNX27, and SNX1) that increase and four (huntingtin-interacting protein 1 [HIP1], HIP14, GASP-1, and Nedd4) that decrease insulin secretion from murine insulinoma MIN6B1 cells in response to the GLP-1 analog exendin-4. The roles of HIP1 and the endosomal SNX1 and SNX27 were further characterized in mouse and human β-cell lines and human islets. While HIP1 was required for the coupling of cell surface GLP-1R activation with clathrin-dependent endocytosis, the SNXs were found to control the balance between GLP-1R plasma membrane recycling and lysosomal degradation and, in doing so, determine the overall β-cell incretin responses. We thus identify key modulators of GLP-1R trafficking and signaling that might provide novel targets to enhance insulin secretion in T2D.
Collapse
Affiliation(s)
- Teresa Buenaventura
- Section of Cell Biology and Functional Genomics and Pancreatic Islet Biology and Diabetes Consortium, Imperial College London, London, U.K
| | - Nisha Kanda
- Section of Cell Biology and Functional Genomics and Pancreatic Islet Biology and Diabetes Consortium, Imperial College London, London, U.K
| | - Phoebe C Douzenis
- Section of Cell Biology and Functional Genomics and Pancreatic Islet Biology and Diabetes Consortium, Imperial College London, London, U.K
| | - Ben Jones
- Section of Investigative Medicine, Imperial College London, London, U.K
| | - Stephen R Bloom
- Section of Investigative Medicine, Imperial College London, London, U.K
| | - Pauline Chabosseau
- Section of Cell Biology and Functional Genomics and Pancreatic Islet Biology and Diabetes Consortium, Imperial College London, London, U.K
| | | | - Domenico Bosco
- Department of Surgery, University of Geneva, Geneva, Switzerland
| | - Lorenzo Piemonti
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Paul R Johnson
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, U.K
| | - A M James Shapiro
- Clinical Islet Laboratory and Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics and Pancreatic Islet Biology and Diabetes Consortium, Imperial College London, London, U.K
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics and Pancreatic Islet Biology and Diabetes Consortium, Imperial College London, London, U.K.
| |
Collapse
|
60
|
Eichel K, von Zastrow M. Subcellular Organization of GPCR Signaling. Trends Pharmacol Sci 2018; 39:200-208. [PMID: 29478570 PMCID: PMC5830169 DOI: 10.1016/j.tips.2017.11.009] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/26/2017] [Accepted: 11/27/2017] [Indexed: 01/09/2023]
Abstract
G protein-coupled receptors (GPCRs) comprise a large and diverse class of signal-transducing receptors that undergo dynamic and isoform-specific membrane trafficking. GPCRs thus have an inherent potential to initiate or regulate signaling reactions from multiple membrane locations. This review discusses emerging insights into the subcellular organization of GPCR function in mammalian cells, focusing on signaling transduced by heterotrimeric G proteins and β-arrestins. We summarize recent evidence indicating that GPCR-mediated activation of G proteins occurs not only from the plasma membrane (PM) but also from endosomes and Golgi membranes and that β-arrestin-dependent signaling can be transduced from the PM by β-arrestin trafficking to clathrin-coated pits (CCPs) after dissociation from a ligand-activated GPCR.
Collapse
Affiliation(s)
- Kelsie Eichel
- Program in Biochemistry and Molecular Biology, University of California, San Francisco School of Medicine, San Francisco, CA 94158, USA
| | - Mark von Zastrow
- Department of Psychiatry, University of California, San Francisco School of Medicine, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, CA 94158, USA.
| |
Collapse
|
61
|
Gorvin CM, Rogers A, Hastoy B, Tarasov AI, Frost M, Sposini S, Inoue A, Whyte MP, Rorsman P, Hanyaloglu AC, Breitwieser GE, Thakker RV. AP2σ Mutations Impair Calcium-Sensing Receptor Trafficking and Signaling, and Show an Endosomal Pathway to Spatially Direct G-Protein Selectivity. Cell Rep 2018; 22:1054-1066. [PMID: 29420171 PMCID: PMC5792449 DOI: 10.1016/j.celrep.2017.12.089] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 11/30/2017] [Accepted: 12/22/2017] [Indexed: 12/29/2022] Open
Abstract
Spatial control of G-protein-coupled receptor (GPCR) signaling, which is used by cells to translate complex information into distinct downstream responses, is achieved by using plasma membrane (PM) and endocytic-derived signaling pathways. The roles of the endomembrane in regulating such pleiotropic signaling via multiple G-protein pathways remain unknown. Here, we investigated the effects of disease-causing mutations of the adaptor protein-2 σ subunit (AP2σ) on signaling by the class C GPCR calcium-sensing receptor (CaSR). These AP2σ mutations increase CaSR PM expression yet paradoxically reduce CaSR signaling. Hypercalcemia-associated AP2σ mutations reduced CaSR signaling via Gαq/11 and Gαi/o pathways. The mutations also delayed CaSR internalization due to prolonged residency time of CaSR in clathrin structures that impaired or abolished endosomal signaling, which was predominantly mediated by Gαq/11. Thus, compartmental bias for CaSR-mediated Gαq/11 endomembrane signaling provides a mechanistic basis for multidimensional GPCR signaling. Disease-causing AP2σ mutants impair Gαq/11 and Gαi/o signaling by CaSR, a class C GPCR AP2σ mutants impair trafficking of the CaSR The CaSR can signal by a sustained endosomal pathway CaSR differentially uses Gαq/11 and Gαi/o for cell-surface and endosomal signaling
Collapse
Affiliation(s)
- Caroline M Gorvin
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
| | - Angela Rogers
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Benoit Hastoy
- Diabetes Research Laboratory, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Andrei I Tarasov
- Diabetes Research Laboratory, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Morten Frost
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Silvia Sposini
- Institute of Reproductive and Developmental Biology, Faculty of Medicine, Imperial College London, London, UK
| | - Asuka Inoue
- Laboratory of Molecular and Cellular Biochemistry, Tohoku University, Sendai, Japan; Japan Science and Technology (JST) Agency, Precursory Research for Embryonic Science and Technology (PRESTO), Kawaguchi, Japan
| | - Michael P Whyte
- Center for Metabolic Bone Disease and Molecular Research, Shriners Hospitals for Children, St. Louis, MO, USA
| | - Patrik Rorsman
- Diabetes Research Laboratory, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Faculty of Medicine, Imperial College London, London, UK
| | - Gerda E Breitwieser
- Geisinger Clinic, Weis Center for Research, Department of Functional and Molecular Genomics, Danville, PA, USA
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
62
|
Evolving View of Membrane Trafficking and Signaling Systems for G Protein-Coupled Receptors. ENDOCYTOSIS AND SIGNALING 2018; 57:273-299. [DOI: 10.1007/978-3-319-96704-2_10] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
63
|
Sayers N, Hanyaloglu AC. Intracellular Follicle-Stimulating Hormone Receptor Trafficking and Signaling. Front Endocrinol (Lausanne) 2018; 9:653. [PMID: 30450081 PMCID: PMC6225286 DOI: 10.3389/fendo.2018.00653] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 10/17/2018] [Indexed: 12/18/2022] Open
Abstract
Models of G protein-coupled receptor (GPCR) signaling have dramatically altered over the past two decades. Indeed, GPCRs such as the follicle-stimulating hormone receptor (FSHR) have contributed to these new emerging models. We now understand that receptor signaling is highly organized at a spatial level, whereby signaling not only occurs from the plasma membrane but distinct intracellular compartments. Recent studies in the role of membrane trafficking and spatial organization of GPCR signaling in regulating gonadotropin hormone receptor activity has identified novel intracellular compartments, which are tightly linked with receptor signaling and reciprocally regulated by the cellular trafficking machinery. Understanding the impact of these cell biological mechanisms to physiology and pathophysiology is emerging for certain GPCRs. However, for FSHR, the potential impact in both health and disease and the therapeutic possibilities of these newly identified systems is currently unknown, but offers the potential to reassess prior strategies, or unveil novel opportunities, in targeting this receptor.
Collapse
|
64
|
Ulloa-Aguirre A, Zariñán T, Jardón-Valadez E, Gutiérrez-Sagal R, Dias JA. Structure-Function Relationships of the Follicle-Stimulating Hormone Receptor. Front Endocrinol (Lausanne) 2018; 9:707. [PMID: 30555414 PMCID: PMC6281744 DOI: 10.3389/fendo.2018.00707] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/09/2018] [Indexed: 12/16/2022] Open
Abstract
The follicle-stimulating hormone receptor (FSHR) plays a crucial role in reproduction. This structurally complex receptor is a member of the G-protein coupled receptor (GPCR) superfamily of membrane receptors. As with the other structurally similar glycoprotein hormone receptors (the thyroid-stimulating hormone and luteinizing hormone-chorionic gonadotropin hormone receptors), the FSHR is characterized by an extensive extracellular domain, where binding to FSH occurs, linked to the signal specificity subdomain or hinge region. This region is involved in ligand-stimulated receptor activation whereas the seven transmembrane domain is associated with receptor activation and transmission of the activation process to the intracellular loops comprised of amino acid sequences, which predicate coupling to effectors, interaction with adapter proteins, and triggering of downstream intracellular signaling. In this review, we describe the most important structural features of the FSHR intimately involved in regulation of FSHR function, including trafficking, dimerization, and oligomerization, ligand binding, agonist-stimulated activation, and signal transduction.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- *Correspondence: Alfredo Ulloa-Aguirre
| | - Teresa Zariñán
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Eduardo Jardón-Valadez
- Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana Unidad Lerma, Lerma, Mexico
| | - Rubén Gutiérrez-Sagal
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - James A. Dias
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, United States
| |
Collapse
|
65
|
Follicle-Stimulating Hormone Receptor: Advances and Remaining Challenges. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:1-58. [DOI: 10.1016/bs.ircmb.2018.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
66
|
Zenko D, Hislop JN. Regulation and trafficking of muscarinic acetylcholine receptors. Neuropharmacology 2017; 136:374-382. [PMID: 29138081 DOI: 10.1016/j.neuropharm.2017.11.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/02/2017] [Accepted: 11/10/2017] [Indexed: 12/31/2022]
Abstract
Fidelity of signal transduction relies on cells expressing the appropriate number of functional receptors. Fluctuation in the total number of muscarinic acetylcholine receptors has been implicated in a range of physiological and pathophysiological processes, and the mechanisms responsible for this regulation represent potential molecular targets for therapeutic intervention. This article will review the current literature on the endocytic trafficking of muscarinic receptors and how knowledge of the trafficking of related receptors might influence future studies. This article is part of the Special Issue entitled 'Neuropharmacology on Muscarinic Receptors'.
Collapse
Affiliation(s)
- Dmitry Zenko
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| | - James N Hislop
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom.
| |
Collapse
|
67
|
Spatial encryption of G protein-coupled receptor signaling in endosomes; Mechanisms and applications. Biochem Pharmacol 2017; 143:1-9. [DOI: 10.1016/j.bcp.2017.04.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/25/2017] [Indexed: 01/14/2023]
|
68
|
Bahouth SW, Nooh MM. Barcoding of GPCR trafficking and signaling through the various trafficking roadmaps by compartmentalized signaling networks. Cell Signal 2017; 36:42-55. [PMID: 28449947 PMCID: PMC5512170 DOI: 10.1016/j.cellsig.2017.04.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 01/08/2023]
Abstract
Proper signaling by G protein coupled receptors (GPCR) is dependent on the specific repertoire of transducing, enzymatic and regulatory kinases and phosphatases that shape its signaling output. Activation and signaling of the GPCR through its cognate G protein is impacted by G protein-coupled receptor kinase (GRK)-imprinted "barcodes" that recruit β-arrestins to regulate subsequent desensitization, biased signaling and endocytosis of the GPCR. The outcome of agonist-internalized GPCR in endosomes is also regulated by sequence motifs or "barcodes" within the GPCR that mediate its recycling to the plasma membrane or retention and eventual degradation as well as its subsequent signaling in endosomes. Given the vast number of diverse sequences in GPCR, several trafficking mechanisms for endosomal GPCR have been described. The majority of recycling GPCR, are sorted out of endosomes in a "sequence-dependent pathway" anchored around a type-1 PDZ-binding module found in their C-tails. For a subset of these GPCR, a second "barcode" imprinted onto specific GPCR serine/threonine residues by compartmentalized kinase networks was required for their efficient recycling through the "sequence-dependent pathway". Mutating the serine/threonine residues involved, produced dramatic effects on GPCR trafficking, indicating that they played a major role in setting the trafficking itinerary of these GPCR. While endosomal SNX27, retromer/WASH complexes and actin were required for efficient sorting and budding of all these GPCR, additional proteins were required for GPCR sorting via the second "barcode". Here we will review recent developments in GPCR trafficking in general and the human β1-adrenergic receptor in particular across the various trafficking roadmaps. In addition, we will discuss the role of GPCR trafficking in regulating endosomal GPCR signaling, which promote biochemical and physiological effects that are distinct from those generated by the GPCR signal transduction pathway in membranes.
Collapse
Affiliation(s)
- Suleiman W Bahouth
- Department of Pharmacology, The University of Tennessee Health Sciences Center, 71 S. Manassas, Memphis, TN 38103, USA.
| | - Mohammed M Nooh
- Department of Biochemistry, Faculty of Pharmacy Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| |
Collapse
|
69
|
Lasič E, Stenovec M, Kreft M, Robinson PJ, Zorec R. Dynamin regulates the fusion pore of endo- and exocytotic vesicles as revealed by membrane capacitance measurements. Biochim Biophys Acta Gen Subj 2017; 1861:2293-2303. [PMID: 28669852 DOI: 10.1016/j.bbagen.2017.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/24/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Dynamin is a multidomain GTPase exhibiting mechanochemical and catalytic properties involved in vesicle scission from the plasmalemma during endocytosis. New evidence indicates that dynamin is also involved in exocytotic release of catecholamines, suggesting the existence of a dynamin-regulated structure that couples endo- to exocytosis. METHODS Thus we here employed high-resolution cell-attached capacitance measurements and super-resolution structured illumination microscopy to directly examine single vesicle interactions with the plasmalemma in cultured rat astrocytes treated with distinct pharmacological modulators of dynamin activity. Fluorescent dextrans and the lipophilic plasmalemmal marker DiD were utilized to monitor uptake and distribution of vesicles in the peri-plasmalemmal space and in the cell cytosol. RESULTS Dynamin inhibition with Dynole™-34-2 and Dyngo™-4a prevented vesicle internalization into the cytosol and decreased fusion pore conductance of vesicles that remained attached to the plasmalemma via a narrow fusion pore that lapsed into a state of repetitive opening and closing - flickering. In contrast, the dynamin activator Ryngo™-1-23 promoted vesicle internalization and favored fusion pore closure by prolonging closed and shortening open fusion pore dwell times. Immunocytochemical staining revealed dextran uptake into dynamin-positive vesicles and increased dextran uptake into Syt4- and VAMP2-positive vesicles after dynamin inhibition, indicating prolonged retention of these vesicles at the plasmalemma. CONCLUSIONS Our results have provided direct evidence for a role of dynamin in regulation of fusion pore geometry and kinetics of endo- and exocytotic vesicles, indicating that both share a common dynamin-regulated structural intermediate, the fusion pore.
Collapse
Affiliation(s)
- Eva Lasič
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Matjaž Stenovec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia; Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia.
| | - Marko Kreft
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia; Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia; University of Ljubljana, Biotechnical Faculty, Department of Biology, CPAE, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Phillip J Robinson
- Children's Medical Research Institute, The University of Sydney, Australia
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia; Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia.
| |
Collapse
|
70
|
Weinberg ZY, Zajac AS, Phan T, Shiwarski DJ, Puthenveedu MA. Sequence-Specific Regulation of Endocytic Lifetimes Modulates Arrestin-Mediated Signaling at the µ Opioid Receptor. Mol Pharmacol 2017; 91:416-427. [PMID: 28153854 PMCID: PMC5363713 DOI: 10.1124/mol.116.106633] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 01/30/2017] [Indexed: 12/26/2022] Open
Abstract
Functional selectivity at the µ opioid receptor (µR), a prototypical G-protein-coupled receptor that is a physiologically relevant target for endogenous opioid neurotransmitters and analgesics, has been a major focus for drug discovery in the recent past. Functional selectivity is a cumulative effect of the magnitudes of individual signaling pathways, e.g., the Gαi-mediated and the arrestin-mediated pathways for µR. The present work tested the hypothesis that lifetimes of agonist-induced receptor-arrestin clusters at the cell surface control the magnitude of arrestin signaling, and therefore functional selectivity, at µR. We show that endomorphin-2 (EM2), an arrestin-biased ligand for µR, lengthens surface lifetimes of receptor-arrestin clusters significantly compared with morphine. The lengthening of lifetimes required two specific leucines on the C-terminal tail of µR. Mutation of these leucines to alanines decreased the magnitude of arrestin-mediated signaling by EM2 without affecting G-protein signaling, suggesting that lengthened endocytic lifetimes were required for arrestin-biased signaling by EM2. Lengthening surface lifetimes by pharmacologically slowing endocytosis was sufficient to increase arrestin-mediated signaling by both EM2 and the clinically relevant agonist morphine. Our findings show that distinct ligands can leverage specific sequence elements on µR to regulate receptor endocytic lifetimes and the magnitude of arrestin-mediated signaling, and implicate these sequences as important determinants of functional selectivity in the opioid system.
Collapse
Affiliation(s)
- Zara Y Weinberg
- Department of Biological Sciences, Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Amanda S Zajac
- Department of Biological Sciences, Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Tiffany Phan
- Department of Biological Sciences, Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Daniel J Shiwarski
- Department of Biological Sciences, Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Manojkumar A Puthenveedu
- Department of Biological Sciences, Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| |
Collapse
|
71
|
Janssens J, Lu D, Ni B, Chadwick W, Siddiqui S, Azmi A, Etienne H, Jushaj A, van Gastel J, Martin B, Maudsley S. Development of Precision Small-Molecule Proneurotrophic Therapies for Neurodegenerative Diseases. VITAMINS AND HORMONES 2016; 104:263-311. [PMID: 28215298 DOI: 10.1016/bs.vh.2016.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Age-related neurodegenerative diseases, such as Alzheimer's disease, will represent one of the largest future burdens on worldwide healthcare systems due to the increasing proportion of elderly in our society. As deficiencies in neurotrophins are implicated in the pathogenesis of many age-related neurodegenerative disorders, it is reasonable to consider that global neurotrophin resistance may also become a major healthcare threat. Central nervous system networks are effectively maintained through aging by neuroprotective and neuroplasticity signaling mechanisms which are predominantly controlled by neurotrophin receptor signaling. Neurotrophin receptors are single pass receptor tyrosine kinases that form dimeric structures upon ligand binding to initiate cellular signaling events that control many protective and plasticity-related pathways. Declining functionality of the neurotrophin ligand-receptor system is considered one of the hallmarks of neuropathological aging. Therefore, it is imperative to develop effective therapeutic strategies to contend with this significant issue. While the therapeutic applications of cognate ligands for neurotrophin receptors are limited, the development of nonpeptidergic, small-molecule ligands can overcome these limitations, and productively regulate this important receptor system with beneficial effects. Using our advanced knowledge of the high-dimensionality complexity of receptor systems, the future generation of precision medicines targeting these systems will be an attainable goal.
Collapse
Affiliation(s)
- J Janssens
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - D Lu
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - B Ni
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - W Chadwick
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Siddiqui
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - A Azmi
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - H Etienne
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - A Jushaj
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - J van Gastel
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - B Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Maudsley
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium; Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States.
| |
Collapse
|
72
|
Bowman SL, Shiwarski DJ, Puthenveedu MA. Distinct G protein-coupled receptor recycling pathways allow spatial control of downstream G protein signaling. J Cell Biol 2016; 214:797-806. [PMID: 27646272 PMCID: PMC5037407 DOI: 10.1083/jcb.201512068] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 08/12/2016] [Indexed: 12/22/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are recycled via a sequence-dependent pathway that is spatially and biochemically distinct from bulk recycling. Why there are two distinct recycling pathways from the endosome is a fundamental question in cell biology. In this study, we show that the separation of these two pathways is essential for normal spatial encoding of GPCR signaling. The prototypical β-2 adrenergic receptor (B2AR) activates Gα stimulatory protein (Gαs) on the endosome exclusively in sequence-dependent recycling tubules marked by actin/sorting nexin/retromer tubular (ASRT) microdomains. B2AR was detected in an active conformation in bulk recycling tubules, but was unable to activate Gαs. Protein kinase A phosphorylation of B2AR increases the fraction of receptors localized to ASRT domains and biases the downstream transcriptional effects of B2AR to genes controlled by endosomal signals. Our results identify the physiological relevance of separating GPCR recycling from bulk recycling and suggest a mechanism to tune downstream responses of GPCR signaling by manipulating the spatial origin of G protein signaling.
Collapse
Affiliation(s)
- Shanna Lynn Bowman
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | | | | |
Collapse
|
73
|
Nooh MM, Mancarella S, Bahouth SW. Identification of novel transplantable GPCR recycling motif for drug discovery. Biochem Pharmacol 2016; 120:22-32. [PMID: 27645110 DOI: 10.1016/j.bcp.2016.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/14/2016] [Indexed: 12/12/2022]
Abstract
β1-Adrenergic receptor (β1-AR) agonists and antagonists are widely used in the treatment of major cardiovascular diseases such as heart failure and hypertension. The β1-AR like other G protein-coupled receptors (GPCRs) are endocytosed in response to intense agonist activation. Recycling of the agonist-internalized β1-AR is dependent on its carboxy-terminal type-1 PSD-95/DLG/ZO1 (PDZ) and on phospho-serine312 in the third intracellular loop of the β1-AR. Progressive elongation of the β1-AR at its C-tail inactivated the PDZ-biding domain and inhibited the recycling of the β1-AR. However, fusing a twenty amino acid peptide derived from the multiple cloning region of the mammalian expression vector pCDNA3 to the C-tail of the β1-AR (β1-AR[+20]) produced a chimeric β1-AR that recycled rapidly and efficiently. The β1-AR[+20] recycled in a type-1 PDZ and phospho-Ser312-independent manner, indicating that this peptide provided a general GPCR recycling signal. Fusing the enhanced yellow fluorescent protein (EYFP) down-stream of β1-AR[+20] generated a β1-AR-EYFP chimera that was expressed on the membrane and recycled efficiently after agonist-induced internalization. This construct trafficked in a PDZ-SNX27/retromer-independent manner. We also fused EYFP to the N-terminus of the β1-AR to created EYFP-WT β1-AR. This construct recycled in PDZ and SNX27/retromer dependent manner. These β1-AR-EYFP constructs would be useful for high throughput screening (HTS) programs to identify new entities that would interfere with the recycling of agonist internalized GPCR that traffic in PDZ-dependent vs. PDZ-independent roadmaps.
Collapse
Affiliation(s)
- Mohammed M Nooh
- Department of Pharmacology, The University of Tennessee Health Sciences Center, 71 S. Manassas, Memphis, TN 38103, USA; Department of Biochemistry, Faculty of Pharmacy Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| | - Salvatore Mancarella
- Department of Physiology, The University of Tennessee Health Sciences Center, 71 S. Manassas, Memphis, TN 38103, USA
| | - Suleiman W Bahouth
- Department of Pharmacology, The University of Tennessee Health Sciences Center, 71 S. Manassas, Memphis, TN 38103, USA.
| |
Collapse
|
74
|
Caldieri G, Sigismund S. Spatial resolution of cAMP signaling by soluble adenylyl cyclase. J Cell Biol 2016; 214:125-7. [PMID: 27402955 PMCID: PMC4949454 DOI: 10.1083/jcb.201606123] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023] Open
Abstract
G protein–coupled receptor signaling starts at the plasma membrane and continues at endosomal stations. In this issue, Inda et al. (2016. J. Cell Biol.http://dx.doi.org/10.1083/jcb.201512075) show that different forms of adenylyl cyclase are activated at the plasma membrane versus endosomes, providing a rationale for the spatial encoding of cAMP signaling.
Collapse
Affiliation(s)
- Giusi Caldieri
- Fondazione Istituto FIRC di Oncologia Molecolare, 20139 Milan, Italy
| | - Sara Sigismund
- Fondazione Istituto FIRC di Oncologia Molecolare, 20139 Milan, Italy
| |
Collapse
|
75
|
Lyga S, Volpe S, Werthmann RC, Götz K, Sungkaworn T, Lohse MJ, Calebiro D. Persistent cAMP Signaling by Internalized LH Receptors in Ovarian Follicles. Endocrinology 2016; 157:1613-21. [PMID: 26828746 DOI: 10.1210/en.2015-1945] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
A crucial event in female reproduction occurs at midcycle, when a LH peak induces the final maturation of ovarian follicles. LH signals via a G protein-coupled receptor selectively expressed in the outermost follicular cell layers. However, how LH signals are relayed inside these cells and finally to the oocyte is incompletely understood. Here, we monitored LH signaling in intact ovarian follicles of transgenic mice expressing a fluorescent cAMP sensor. We found that LH stimulation induces 2 phases of cAMP signaling in all cell layers surrounding the oocyte. Interfering with LH receptor internalization abolished the second, persistent cAMP phase and partially inhibited oocyte meiosis resumption. These data suggest that persistent cAMP signals from internalized LH receptors contribute to transmitting LH effects inside follicle cells and ultimately to the oocyte. Thus, this study indicates that the recently proposed paradigm of cAMP signaling by internalized G protein-coupled receptors is implicated in receptor function and is physiologically relevant.
Collapse
Affiliation(s)
- Sandra Lyga
- Institute of Pharmacology and Toxicology, University of Würzburg, and Rudolf Virchow Center, Bio-Imaging Center, 97078 Würzburg, Germany
| | - Silvia Volpe
- Institute of Pharmacology and Toxicology, University of Würzburg, and Rudolf Virchow Center, Bio-Imaging Center, 97078 Würzburg, Germany
| | - Ruth C Werthmann
- Institute of Pharmacology and Toxicology, University of Würzburg, and Rudolf Virchow Center, Bio-Imaging Center, 97078 Würzburg, Germany
| | - Konrad Götz
- Institute of Pharmacology and Toxicology, University of Würzburg, and Rudolf Virchow Center, Bio-Imaging Center, 97078 Würzburg, Germany
| | - Titiwat Sungkaworn
- Institute of Pharmacology and Toxicology, University of Würzburg, and Rudolf Virchow Center, Bio-Imaging Center, 97078 Würzburg, Germany
| | - Martin J Lohse
- Institute of Pharmacology and Toxicology, University of Würzburg, and Rudolf Virchow Center, Bio-Imaging Center, 97078 Würzburg, Germany
| | - Davide Calebiro
- Institute of Pharmacology and Toxicology, University of Würzburg, and Rudolf Virchow Center, Bio-Imaging Center, 97078 Würzburg, Germany
| |
Collapse
|
76
|
Vishwanatha KS, Bäck N, Lam TT, Mains RE, Eipper BA. O-Glycosylation of a Secretory Granule Membrane Enzyme Is Essential for Its Endocytic Trafficking. J Biol Chem 2016; 291:9835-50. [PMID: 26961877 DOI: 10.1074/jbc.m115.711838] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Indexed: 01/08/2023] Open
Abstract
Peptidylglycine α-amidating monooxygenase (PAM) (EC 1.14.17.3) catalyzes peptide amidation, a crucial post-translational modification, through the sequential actions of its monooxygenase (peptidylglycine α-hydroxylating monooxygenase) and lyase (peptidyl-α-hydroxyglycine α-amidating lyase (PAL)) domains. Alternative splicing generates two different regions that connect the protease-resistant catalytic domains. Inclusion of exon 16 introduces a pair of Lys residues, providing a site for controlled endoproteolytic cleavage of PAM and the separation of soluble peptidylglycine α-hydroxylating monooxygenase from membrane-associated PAL. Exon 16 also includes two O-glycosylation sites. PAM-1 lacking both glycosylation sites (PAM-1/OSX; where OSX is O-glycan-depleted mutant of PAM-1) was stably expressed in AtT-20 corticotrope tumor cells. In PAM-1/OSX, a cleavage site for furin-like convertases was exposed, generating a shorter form of membrane-associated PAL. The endocytic trafficking of PAM-1/OSX differed dramatically from that of PAM-1. A soluble fragment of the cytosolic domain of PAM-1 was produced in the endocytic pathway and entered the nucleus; very little soluble fragment of the cytosolic domain was produced from PAM-1/OSX. Internalized PAM-1/OSX was rapidly degraded; unlike PAM-1, very little internalized PAM-1/OSX was detected in multivesicular bodies. Blue native PAGE analysis identified high molecular weight complexes containing PAM-1; the ability of PAM-1/OSX to form similar complexes was markedly diminished. By promoting the formation of high molecular weight complexes, O-glycans may facilitate the recycling of PAM-1 through the endocytic compartment.
Collapse
Affiliation(s)
| | - Nils Bäck
- the Department of Anatomy, Faculty of Medicine, University of Helsinki, Fin-00014, Helsinki, Finland, and
| | - TuKiet T Lam
- the W. M. Keck Foundation Biotechnology Resource Laboratory, Yale/Keck MS and Proteomics Resource, Yale/NIDA Neuroproteomics Center, Yale University, New Haven, Connecticut 06511
| | | | - Betty A Eipper
- From the Departments of Neuroscience and Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, Connecticut 06030,
| |
Collapse
|
77
|
Specific cell surface labeling of GPCRs using split GFP. Sci Rep 2016; 6:20568. [PMID: 26857153 PMCID: PMC4746647 DOI: 10.1038/srep20568] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/07/2016] [Indexed: 01/13/2023] Open
Abstract
Specific cell surface labeling is essential for visualizing the internalization processes of G-protein coupled receptors (GPCRs) and for gaining mechanistic insight of GPCR functions. Here we present a rapid, specific, and versatile labeling scheme for GPCRs at living-cell membrane with the use of a split green fluorescent protein (GFP). Demonstrated with two GPCRs, GPR17 and CysLT2R, we show that two β-stands (β-stands 10 and 11) derived from a superfolder GFP (sfGFP) can be engineered to one of the three extracellular loop of a GPCR. The complementary fragment of sfGFP has nine β-strands (β-stands 1-9) that carries the mature fluorophore, and can be proteolytically derived from the full-length sfGFP. Separately the GFP fragments are non-fluorescent, but become fluorescent upon assembly, thus allowing specific labeling of the target proteins. The two GFP fragments rapidly assemble and the resulting complex is extremely tight under non-denaturing conditions, which allows real-time and quantitative assessment of the internalized GPCRs. We envision that this labeling scheme will be of great use for labeling other membrane proteins in various biological and pharmacological applications.
Collapse
|
78
|
REPLACR-mutagenesis, a one-step method for site-directed mutagenesis by recombineering. Sci Rep 2016; 6:19121. [PMID: 26750263 PMCID: PMC4707547 DOI: 10.1038/srep19121] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 12/04/2015] [Indexed: 12/22/2022] Open
Abstract
Mutagenesis is an important tool to study gene regulation, model disease-causing mutations and for functional characterisation of proteins. Most of the current methods for mutagenesis involve multiple step procedures. One of the most accurate methods for genetically altering DNA is recombineering, which uses bacteria expressing viral recombination proteins. Recently, the use of in vitro seamless assembly systems using purified enzymes for multiple-fragment cloning as well as mutagenesis is gaining ground. Although these in vitro isothermal reactions are useful when cloning multiple fragments, for site-directed mutagenesis it is unnecessary. Moreover, the use of purified enzymes in vitro is not only expensive but also more inaccurate than the high-fidelity recombination inside bacteria. Here we present a single-step method, named REPLACR-mutagenesis (Recombineering of Ends of linearised PLAsmids after PCR), for creating mutations (deletions, substitutions and additions) in plasmids by in vivo recombineering. REPLACR-mutagenesis only involves transformation of PCR products in bacteria expressing Red/ET recombineering proteins. Modifications in a variety of plasmids up to bacterial artificial chromosomes (BACs; 144 kb deletion) have been achieved by this method. The presented method is more robust, involves fewer steps and is cost-efficient.
Collapse
|
79
|
Irannejad R, Tsvetanova NG, Lobingier BT, von Zastrow M. Effects of endocytosis on receptor-mediated signaling. Curr Opin Cell Biol 2015; 35:137-43. [PMID: 26057614 PMCID: PMC4529812 DOI: 10.1016/j.ceb.2015.05.005] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/18/2015] [Accepted: 05/20/2015] [Indexed: 01/31/2023]
Abstract
Cellular mechanisms of membrane traffic and signal transduction are deeply interconnected. The present review discusses how membrane trafficking in the endocytic pathway impacts receptor-mediated signaling. Examples of recent progress are highlighted, focusing on the endocytosis-signaling nexus in mammals.
Collapse
Affiliation(s)
- Roshanak Irannejad
- Departments of Psychiatry and of Cellular & Molecular Pharmacology, University of California, San Francisco School of Medicine, 600 16th Street, San Francisco CA 94158-2140 USA
| | - Nikoleta G. Tsvetanova
- Departments of Psychiatry and of Cellular & Molecular Pharmacology, University of California, San Francisco School of Medicine, 600 16th Street, San Francisco CA 94158-2140 USA
| | - Braden T. Lobingier
- Departments of Psychiatry and of Cellular & Molecular Pharmacology, University of California, San Francisco School of Medicine, 600 16th Street, San Francisco CA 94158-2140 USA
| | - Mark von Zastrow
- Departments of Psychiatry and of Cellular & Molecular Pharmacology, University of California, San Francisco School of Medicine, 600 16th Street, San Francisco CA 94158-2140 USA
| |
Collapse
|
80
|
West C, Hanyaloglu AC. Minireview: Spatial Programming of G Protein-Coupled Receptor Activity: Decoding Signaling in Health and Disease. Mol Endocrinol 2015; 29:1095-106. [PMID: 26121235 DOI: 10.1210/me.2015-1065] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Probing the multiplicity of hormone signaling via G protein-coupled receptors (GPCRs) has demonstrated the complex signal pathways that underlie the multiple functions these receptors play in vivo. This is highly pertinent for the GPCRs key in reproduction and pregnancy that are exposed to cyclical and dynamic changes in their extracellular milieu. How such functional pleiotropy in GPCR signaling is translated to specific downstream cellular responses, however, is largely unknown. Emerging data strongly support mechanisms for a central role of receptor location in signal regulation via membrane trafficking. In this review, we discuss current progress in our understanding of the role membrane trafficking plays in location control of GPCR signaling, from organized plasma membrane signaling microdomains, potentially provided by both distinct endocytic and exocytic pathways, to more recent evidence for spatial control within the endomembrane system. Application of these emerging mechanisms in their relevance to GPCR activity in physiological and pathophysiological conditions will also be discussed, and in improving therapeutic strategies that exploits these mechanisms in order to program highly regulated and distinct signaling profiles.
Collapse
Affiliation(s)
- Camilla West
- Institute of Reproductive Biology and Development, Department of Surgery and Cancer, Imperial College London, London, W12 0NN, United Kingdom
| | - Aylin C Hanyaloglu
- Institute of Reproductive Biology and Development, Department of Surgery and Cancer, Imperial College London, London, W12 0NN, United Kingdom
| |
Collapse
|
81
|
Walther C, Ferguson SSG. Minireview: Role of intracellular scaffolding proteins in the regulation of endocrine G protein-coupled receptor signaling. Mol Endocrinol 2015; 29:814-30. [PMID: 25942107 DOI: 10.1210/me.2015-1091] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The majority of hormones stimulates and mediates their signal transduction via G protein-coupled receptors (GPCRs). The signal is transmitted into the cell due to the association of the GPCRs with heterotrimeric G proteins, which in turn activates an extensive array of signaling pathways to regulate cell physiology. However, GPCRs also function as scaffolds for the recruitment of a variety of cytoplasmic protein-interacting proteins that bind to both the intracellular face and protein interaction motifs encoded by GPCRs. The structural scaffolding of these proteins allows GPCRs to recruit large functional complexes that serve to modulate both G protein-dependent and -independent cellular signaling pathways and modulate GPCR intracellular trafficking. This review focuses on GPCR interacting PSD95-disc large-zona occludens domain containing scaffolds in the regulation of endocrine receptor signaling as well as their potential role as therapeutic targets for the treatment of endocrinopathies.
Collapse
Affiliation(s)
- Cornelia Walther
- J. Allyn Taylor Centre for Cell Biology (C.W., S.S.G.F.), Robarts Research Institute, and Department of Physiology and Pharmacology (S.S.G.F.), University of Western Ontario, London, Ontario, Canada N6A 5K8
| | - Stephen S G Ferguson
- J. Allyn Taylor Centre for Cell Biology (C.W., S.S.G.F.), Robarts Research Institute, and Department of Physiology and Pharmacology (S.S.G.F.), University of Western Ontario, London, Ontario, Canada N6A 5K8
| |
Collapse
|
82
|
Mörl K, Beck-Sickinger AG. Intracellular Trafficking of Neuropeptide Y Receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:73-96. [PMID: 26055055 DOI: 10.1016/bs.pmbts.2015.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The multireceptor multiligand system of neuropeptide Y receptors and their ligands is involved in the regulation of a multitude of physiological and pathophysiological processes. Specific expression patterns, ligand-binding modes, and signaling properties contribute to the complex network regulating distinct cellular responses. Intracellular trafficking processes are important key steps that are regulated in context with accessory proteins. These proteins exert their influence by interacting directly or indirectly with the receptors, causing modification of the receptors, or operating as scaffolds for the assembly of larger signaling complexes. On the intracellular receptor faces, sequence-specific motifs have been identified that play an important role in this process. Interestingly, it is also possible to influence the receptor internalization by modification of the peptide ligand.
Collapse
Affiliation(s)
- Karin Mörl
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Universität Leipzig, Leipzig, Germany.
| | - Annette G Beck-Sickinger
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Universität Leipzig, Leipzig, Germany
| |
Collapse
|
83
|
Burczyk M, Burkhalter MD, Blätte T, Matysik S, Caron MG, Barak LS, Philipp M. Phenotypic regulation of the sphingosine 1-phosphate receptor miles apart by G protein-coupled receptor kinase 2. Biochemistry 2015; 54:765-75. [PMID: 25555130 PMCID: PMC4310627 DOI: 10.1021/bi501061h] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
The evolutionarily conserved DRY
motif at the end of the third
helix of rhodopsin-like, class-A G protein-coupled receptors (GPCRs)
is a major regulator of receptor stability, signaling activity, and
β-arrestin-mediated internalization. Substitution of the DRY
arginine with histidine in the human vasopressin receptor results
in a loss-of-function phenotype associated with diabetes insipidus.
The analogous R150H substitution of the DRY motif in zebrafish sphingosine-1
phosphate receptor 2 (S1p2) produces a mutation, miles apart m93 (milm93), that not only disrupts signaling but
also impairs heart field migration. We hypothesized that constitutive
S1p2 desensitization is the underlying cause of this strong zebrafish
developmental defect. We observed in cell assays that the wild-type
S1p2 receptor is at the cell surface whereas in distinct contrast
the S1p2 R150H receptor is found in intracellular vesicles, blocking
G protein but not arrestin signaling activity. Surface S1p2 R150H
expression could be restored by inhibition of G protein-coupled receptor
kinase 2 (GRK2). Moreover, we observed that β-arrestin 2 and
GRK2 colocalize with S1p2 in developing zebrafish embryos and depletion
of GRK2 in the S1p2 R150H miles apart zebrafish partially rescued
cardia bifida. The ability of reduced GRK2 activity to reverse a developmental
phenotype associated with constitutive desensitization supports efforts
to genetically or pharmacologically target this kinase in diseases
involving biased GPCR signaling.
Collapse
Affiliation(s)
- Martina Burczyk
- Institute for Biochemistry and Molecular Biology, Ulm University , 89081 Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
84
|
Ayoub MA, Landomiel F, Gallay N, Jégot G, Poupon A, Crépieux P, Reiter E. Assessing Gonadotropin Receptor Function by Resonance Energy Transfer-Based Assays. Front Endocrinol (Lausanne) 2015; 6:130. [PMID: 26379624 PMCID: PMC4550792 DOI: 10.3389/fendo.2015.00130] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/10/2015] [Indexed: 11/13/2022] Open
Abstract
Gonadotropin receptors belong to the super family of G protein-coupled receptors and mediate the physiological effects of follicle-stimulating hormone (FSHR) and luteinizing hormone (LHR). Their central role in the control of reproductive function has made them the focus of intensive studies. Upon binding to their cognate hormone, they trigger complex signaling and trafficking mechanisms that are tightly regulated in concentration, time, and space. Classical cellular assays often fail to capture all these dynamics. Here, we describe the use of various bioluminescence and fluorescence resonance energy transfer (BRET and FRET) assays to investigate the activation and regulation of FSHR and LHR in real-time, in living cells (i.e., transiently expressed in human embryonic kidney 293 cells). Indeed, the dynamics of hormone-mediated heterotrimeric G protein activation, cyclic adenosine-monophosphate (cAMP) production, calcium release, β-arrestin 2 recruitment, and receptor internalization/recycling was assessed. Kinetics and dose-response analyses confirmed the expected pharmacological and signaling properties of hFSHR and hLHR but revealed interesting characteristics when considering the two major pathways (cAMP and β-arrestin 2) of the two receptors assessed by BRET. Indeed, the EC50 values were in picomolar range for cAMP production while nanomolar range was observed for β-arrestin 2 recruitment as well as receptor internalization. Interestingly, the predicted receptor occupancy indicates that the maximal G protein activation and cAMP response occur at <10% of receptor occupancy whereas >90% of activated receptors is required to achieve full β-arrestin 2 recruitment and subsequent receptor internalization. The rapid receptor internalization was also followed by a recycling phase. Collectively, our data reveal that β-arrestin-mediated desensitization, internalization, and the subsequent fast recycling of receptors at the plasma membrane may provide a mechanistic ground to the "spare receptor" paradigm. More generally, the novel tools described here will undoubtedly provide the scientific community investigating gonadotropin receptors with powerful means to decipher their pharmacology and signaling with the prospect of pathophysiological and drug discovery applications.
Collapse
Affiliation(s)
- Mohammed Akli Ayoub
- Biologie et Bioinformatique des Systèmes de Signalisation (BIOS) Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- L’Institut français du cheval et de l’équitation (IFCE), Nouzilly, France
- LE STUDIUM Loire Valley Institute for Advanced Studies, Orléans, France
- *Correspondence: Mohammed Akli Ayoub, Institut National de la Recherche Agronomique (INRA) UMR85, CNRS-Université François Rabelais UMR7247, Physiologie de la Reproduction et des Comportements (PRC) - Nouzilly 37380, France,
| | - Flavie Landomiel
- Biologie et Bioinformatique des Systèmes de Signalisation (BIOS) Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- L’Institut français du cheval et de l’équitation (IFCE), Nouzilly, France
| | - Nathalie Gallay
- Biologie et Bioinformatique des Systèmes de Signalisation (BIOS) Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- L’Institut français du cheval et de l’équitation (IFCE), Nouzilly, France
| | - Gwenhael Jégot
- Biologie et Bioinformatique des Systèmes de Signalisation (BIOS) Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- L’Institut français du cheval et de l’équitation (IFCE), Nouzilly, France
| | - Anne Poupon
- Biologie et Bioinformatique des Systèmes de Signalisation (BIOS) Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- L’Institut français du cheval et de l’équitation (IFCE), Nouzilly, France
| | - Pascale Crépieux
- Biologie et Bioinformatique des Systèmes de Signalisation (BIOS) Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- L’Institut français du cheval et de l’équitation (IFCE), Nouzilly, France
| | - Eric Reiter
- Biologie et Bioinformatique des Systèmes de Signalisation (BIOS) Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- L’Institut français du cheval et de l’équitation (IFCE), Nouzilly, France
| |
Collapse
|
85
|
Xu H, Chen H, Li J, Zhao Y, Ghishan FK. Disruption of NHE8 expression impairs Leydig cell function in the testes. Am J Physiol Cell Physiol 2014; 308:C330-8. [PMID: 25472965 DOI: 10.1152/ajpcell.00289.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple sodium/hydrogen exchanger (NHE) isoforms are expressed in the testes, and they play various roles in cell volume regulation, intracellular pH regulation, and fluid absorption. NHE8, the most recently characterized NHE family member, is detected in the Leydig cells in humans and mice in great abundance by immunohistochemistry in the current study. Male mice lacking NHE8 expression were infertile. Despite having intact male reproductive organs, male NHE8-/- mice have smaller testes and lacked spermatozoon in the seminiferous tubules and the epididymis. At the age of 39 wk, few spermogonia were seen in the testis in NHE8-/- mice. Although male NHE8-/- mice have normal serum levels of luteinizing hormone and follicle-stimulating hormone, serum testosterone level was significantly reduced. These mice have decreased expression of luteinizing hormone receptor in the testes. In NHE8 small-interfering RNA-transfected mouse Leydig cells (MLTC-1), silencing of NHE8 decreased the expression of luteinizing hormone receptor by ∼70%. Moreover, loss of NHE8 function in Leydig cells resulted in disorganized luteinizing hormone receptor membrane distribution. Therefore, male infertility in NHE8-/- mice is at least partially due to the disruption of luteinizing hormone receptor distribution and consequent low testosterone production, which leads to Sertoli cell dysfunction. Our work identified a novel role of NHE8 in male fertility through its effect on modifying luteinizing hormone receptor function.
Collapse
Affiliation(s)
- Hua Xu
- Department of Pediatrics, Steel Children's Research Center, The University of Arizona, Tucson, Arizona
| | - Huacong Chen
- Department of Pediatrics, Steel Children's Research Center, The University of Arizona, Tucson, Arizona
| | - Jing Li
- Department of Pediatrics, Steel Children's Research Center, The University of Arizona, Tucson, Arizona
| | - Yang Zhao
- Department of Pediatrics, Steel Children's Research Center, The University of Arizona, Tucson, Arizona
| | - Fayez K Ghishan
- Department of Pediatrics, Steel Children's Research Center, The University of Arizona, Tucson, Arizona
| |
Collapse
|
86
|
Gupta A, Fujita W, Gomes I, Bobeck E, Devi LA. Endothelin-converting enzyme 2 differentially regulates opioid receptor activity. Br J Pharmacol 2014; 172:704-19. [PMID: 24990314 DOI: 10.1111/bph.12833] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 06/17/2014] [Accepted: 06/24/2014] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Opioid receptor function is modulated by post-activation events such as receptor endocytosis, recycling and/or degradation. While it is generally understood that the peptide ligand gets co-endocytosed with the receptor, relatively few studies have investigated the role of the endocytosed peptide and peptide processing enzymes in regulating receptor function. In this study, we focused on endothelin-converting enzyme 2 (ECE2), a member of the neprilysin family of metallopeptidases that exhibits an acidic pH optimum, localizes to an intracellular compartment and selectively processes neuropeptides including opioid peptides in vitro, and examined its role in modulating μ receptor recycling and resensitization. EXPERIMENTAL APPROACH The effect of ECE2 inhibition on hydrolysis of the endocytosed peptide was examined using thin-layer chromatography and on μ opioid receptor trafficking using either elisa or microscopy. The effect of ECE2 inhibition on receptor signalling was measured using a cAMP assay and, in vivo, on antinociception induced by intrathecally administered opioids by the tail-flick assay. KEY RESULTS The highly selective ECE2 inhibitor, S136492, significantly impaired μ receptor recycling and signalling by only those ligands that are ECE2 substrates and this was seen both in heterologous cells and in cells endogenously co-expressing μ receptors with ECE2. We also found that ECE2 inhibition attenuated antinociception mediated only by opioid peptides that are ECE2 substrates. CONCLUSIONS AND IMPLICATIONS These results suggest that ECE2, by selectively processing endogenous opioid peptides in the endocytic compartment, plays a role in modulating opioid receptor activity. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- A Gupta
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | |
Collapse
|