1
|
Matthees ESF, Kletzin LE, Löbbert A, Hoffmann JS, Große C, Gossert AD, Hoffmann C. Isoprenaline shows unique kinase dependencies in stimulating β 1AR-β-arrestin2 interaction compared to endogenous catecholamines. Mol Pharmacol 2025; 107:100041. [PMID: 40354729 DOI: 10.1016/j.molpha.2025.100041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/21/2025] [Accepted: 04/12/2025] [Indexed: 05/14/2025] Open
Abstract
The β1-adrenergic receptor (β1AR) is an essential G protein-coupled receptor in the heart. Its dysregulation represents a hallmark of cardiac diseases. Studies have identified a unique mode of β-arrestin interaction, where β1AR briefly engages with β-arrestins before catalytically accumulating them at the plasma membrane (PM) independently of the receptor. Although receptor phosphorylation crucially impacts β-arrestins, the contributions of specific kinases vital in β1AR regulation remain unclear. Here, we employed G protein-coupled receptor kinase (GRK) GRK2/3/5/6 knockout cells and the protein kinase A inhibitor H89 in bioluminescence resonance energy transfer-based assays to systematically assess GRKs and protein kinase A in direct β-arrestin2 recruitment to β1AR and β-arrestin2 translocation to the PM. Furthermore, we compared the effects of the synthetic agonist isoprenaline with the endogenous catecholamines: epinephrine and norepinephrine. We observed pronounced differences in their kinase dependencies to mediate β-arrestin2 translocation to the PM. Upon isoprenaline stimulation, GRKs strongly influenced β-arrestin2 translocation to the PM but had no effect on direct β-arrestin2 recruitment to β1AR. Additionally, in a GRK2-specific context, protein kinase A inhibition primarily reduced the efficacy of isoprenaline for β-arrestin2 translocation, whereas for GRK5, it decreased potency. Strikingly, these kinase-dependent effects were absent for epinephrine and norepinephrine, suggesting distinct underlying molecular mechanisms for β-arrestin2 accumulation at the PM. This observation could be explained by agonist-specific differences in receptor conformational rearrangements, as suggested by distinct changes in the NMR spectra of β1AR. Our findings highlight that synthetic and endogenous ligands induce distinct molecular mechanisms in β1AR regulation, emphasizing the need to consider these differences when translating molecular insights into physiological contexts. SIGNIFICANCE STATEMENT: Our findings reveal mechanistic differences in β1-adrenergic receptor-mediated catalytic activation of β-arrestin2 by synthetic and endogenous agonists, driven by distinct G protein-coupled receptor kinases and protein kinase A dependencies. Although β-arrestin2 translocation to the PM occurred to similar extents with isoprenaline, epinephrine, and norepinephrine, kinase involvement was crucial only upon Iso stimulation of β1-adrenergic receptor. By elucidating these ligand-specific pathways, this study advances our understanding of β1-adrenergic receptor signaling and regulation while additionally highlighting the importance of considering these differences when translating molecular insights into pathophysiological contexts.
Collapse
Affiliation(s)
- Edda S F Matthees
- Institute for Molecular Cell Biology, Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, Jena, Germany.
| | - Luca E Kletzin
- Institute for Molecular Cell Biology, Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, Jena, Germany
| | - Arnelle Löbbert
- Department of Biology, Institute of Biochemistry, ETH Zürich, Zürich, Switzerland
| | - Jana S Hoffmann
- Institute for Molecular Cell Biology, Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, Jena, Germany
| | - Carolin Große
- Institute for Molecular Cell Biology, Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, Jena, Germany
| | - Alvar D Gossert
- Department of Biology, Institute of Biochemistry, ETH Zürich, Zürich, Switzerland
| | - Carsten Hoffmann
- Institute for Molecular Cell Biology, Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, Jena, Germany.
| |
Collapse
|
2
|
Anderson PJ, Xiao P, Zhong Y, Kaakati A, Alfonso-DeSouza J, Zhang T, Zhang C, Yu K, Qi L, Ding W, Liu S, Pani B, Krishnan A, Chen O, Jassal C, Strawn J, Sun JP, Rajagopal S. β-Arrestin Condensates Regulate G Protein-Coupled Receptor Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.05.647240. [PMID: 40236194 PMCID: PMC11996538 DOI: 10.1101/2025.04.05.647240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
G protein-coupled receptors (GPCRs) are the largest class of receptors in the genome and control many signaling cascades essential for survival. GPCR signaling is regulated by β-arrestins, multifunctional adapter proteins that direct receptor desensitization, internalization, and signaling. While at many GPCRs, β-arrestins interact with a wide array of signaling effectors, it is unclear how β-arrestins promote such varied functions. Here we show that β-arrestins undergo liquid-liquid phase separation (LLPS) to form condensates that regulate GPCR function. We demonstrate that β-arrestin oligomerization occurs in proximity to the GPCR and regulates GPCR functions such as internalization and signaling. This model is supported by a cryoEM structure of the adhesion receptor ADGRE1 in a 2:2 complex with β-arrestin 1, with a β-arrestin orientation that can promote oligomerization. Our work provides a paradigm for β-arrestin condensates as regulators of GPCR function, with LLPS serving as an important promoter of signaling compartmentalization at GPCRs.
Collapse
|
3
|
Fallon BS, Rondem KE, Mumby EJ, English JG. Biased Signaling in G Protein-Coupled Receptors: Understanding the Biological Relevance and Tools for Probing Functionally Selective Ligands. Biochemistry 2025; 64:1425-1436. [PMID: 40100969 DOI: 10.1021/acs.biochem.4c00871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Biased signaling has transformed pharmacology by revealing that receptors, particularly G protein-coupled receptors (GPCRs), can activate specific intracellular pathways selectively rather than uniformly. This discovery enables the development of targeted therapeutics that minimize side effects by precisely modulating receptor activity. Functionally selective ligands, which preferentially activate distinct signaling branches, have become essential tools for exploring receptor mechanisms and uncovering the complexities of GPCR signaling. These ligands help clarify receptor function in various physiological and pathological contexts, offering profound implications for therapeutic innovation. GPCRs, which mediate a wide range of cellular responses through coupling to G proteins and arrestins, are key pharmacological targets, with nearly a third of FDA-approved drugs acting on them. Recent advancements in biosensor development, multiplex assay platforms, and deep mutational scanning methods are improving our ability to define GPCR signaling, allowing for a better understanding of biased signaling pathways.
Collapse
Affiliation(s)
- Braden S Fallon
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84132-2101, United States
| | - Kathleen E Rondem
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84132-2101, United States
| | - Elizabeth J Mumby
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84132-2101, United States
| | - Justin G English
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84132-2101, United States
| |
Collapse
|
4
|
Pacheco J, Peña KA, Savransky S, Gidon A, Hammond GRV, Janetzko J, Vilardaga JP. Fast-diffusing receptor collisions with slow-diffusing peptide ligand assemble the ternary parathyroid hormone-GPCR-arrestin complex. Nat Commun 2024; 15:10499. [PMID: 39627206 PMCID: PMC11615292 DOI: 10.1038/s41467-024-54772-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
The assembly of a peptide ligand, its receptor, and β-arrestin (βarr) into a ternary complex within the cell membrane is a crucial aspect of G protein-coupled receptor (GPCR) signaling. We explore this assembly by attaching fluorescent moieties to the parathyroid hormone (PTH) type 1 receptor (PTH1R), using PTH as a prototypical peptide hormone, along with βarr and clathrin, and recording dual-color single-molecule imaging at the plasma membrane of live cells. Here we show that PTH1R exhibits a near-Brownian diffusion, whereas unbound hormone displays limited mobility and slow lateral diffusion at the cell surface. The formation of the PTH-PTH1R-βarr complex occurs in three sequential steps: (1) receptor and ligand collisions, (2) phosphoinositide (PIP3)-dependent recruitment and conformational change of βarr molecules at the plasma membrane, and (3) collision of most βarr molecules with the ligand-bound receptor within clathrin clusters. Our results elucidate the non-random pathway by which PTH-PTH1R-βarr complex is formed and unveil the critical role of PIP3 in regulating GPCR signaling.
Collapse
Affiliation(s)
- Jonathan Pacheco
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Karina A Peña
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Sofya Savransky
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
- Graduate Program in Molecular Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Alexandre Gidon
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
- Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - John Janetzko
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jean-Pierre Vilardaga
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
- Graduate Program in Molecular Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
- U.S. Department of Veterans Affairs, Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA.
| |
Collapse
|
5
|
Tóth AD, Turu G, Hunyady L. Functional consequences of spatial, temporal and ligand bias of G protein-coupled receptors. Nat Rev Nephrol 2024; 20:722-741. [PMID: 39039165 DOI: 10.1038/s41581-024-00869-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 07/24/2024]
Abstract
G protein-coupled receptors (GPCRs) regulate every aspect of kidney function by mediating the effects of various endogenous and exogenous substances. A key concept in GPCR function is biased signalling, whereby certain ligands may selectively activate specific pathways within the receptor's signalling repertoire. For example, different agonists may induce biased signalling by stabilizing distinct active receptor conformations - a concept that is supported by advances in structural biology. However, the processes underlying functional selectivity in receptor signalling are extremely complex, involving differences in subcellular compartmentalization and signalling dynamics. Importantly, the molecular mechanisms of spatiotemporal bias, particularly its connection to ligand binding kinetics, have been detailed for GPCRs critical to kidney function, such as the AT1 angiotensin receptor (AT1R), V2 vasopressin receptor (V2R) and the parathyroid hormone 1 receptor (PTH1R). This expanding insight into the multifaceted nature of biased signalling paves the way for innovative strategies for targeting GPCR functions; the development of novel biased agonists may represent advanced pharmacotherapeutic approaches to the treatment of kidney diseases and related systemic conditions, such as hypertension, diabetes and heart failure.
Collapse
MESH Headings
- Humans
- Ligands
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/physiology
- Receptors, Vasopressin/metabolism
- Receptors, Vasopressin/physiology
- Animals
- Receptor, Parathyroid Hormone, Type 1/metabolism
- Receptor, Parathyroid Hormone, Type 1/physiology
- Kidney Diseases/metabolism
- Kidney/metabolism
Collapse
Affiliation(s)
- András D Tóth
- Institute of Molecular Life Sciences, Centre of Excellence of the Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - Gábor Turu
- Institute of Molecular Life Sciences, Centre of Excellence of the Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - László Hunyady
- Institute of Molecular Life Sciences, Centre of Excellence of the Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
6
|
Wei X, Wu D, Li J, Wu M, Li Q, Che Z, Cheng X, Cheng Q, Yin F, Zhang H, Wang X, Abtahi S, Zuo L, Hang L, Ma L, Kuo WT, Liu X, Turner JR, Wang H, Xiao J, Wang F. Myeloid beta-arrestin 2 depletion attenuates metabolic dysfunction-associated steatohepatitis via the metabolic reprogramming of macrophages. Cell Metab 2024; 36:2281-2297.e7. [PMID: 39305895 DOI: 10.1016/j.cmet.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/27/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024]
Abstract
Macrophage-mediated inflammation has been implicated in the pathogenesis of metabolic dysfunction-associated steatohepatitis (MASH); however, the immunometabolic program underlying the regulation of macrophage activation remains unclear. Beta-arrestin 2, a multifunctional adaptor protein, is highly expressed in bone marrow tissues and macrophages and is involved in metabolism disorders. Here, we observed that β-arrestin 2 expression was significantly increased in the liver macrophages and circulating monocytes of patients with MASH compared with healthy controls and positively correlated with the severity of metabolic dysfunction-associated steatotic liver disease (MASLD). Global or myeloid Arrb2 deficiency prevented the development of MASH in mice. Further study showed that β-arrestin 2 acted as an adaptor protein and promoted ubiquitination of immune responsive gene 1 (IRG1) to prevent increased itaconate production in macrophages, which resulted in enhanced succinate dehydrogenase activity, thereby promoting the release of mitochondrial reactive oxygen species and M1 polarization. Myeloid β-arrestin 2 depletion may be a potential approach for MASH.
Collapse
Affiliation(s)
- Xiaoli Wei
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dongqing Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Li
- College of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Miaomiao Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; College of Pharmacy, Anhui Medical University, Hefei, China
| | - Qianhui Li
- Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xu Cheng
- Department of Cardiology, First Affiliated Hospital, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Qianying Cheng
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fan Yin
- Department of Pharmacy, Huainan First People's Hospital, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan, China
| | - Hao Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuefu Wang
- College of Pharmacy, Anhui Medical University, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Shabnam Abtahi
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Li Zuo
- School of Basic Medical Sciences, Molecular Biology Laboratory, Anhui Medical University, Hefei, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - Lei Hang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - Lili Ma
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - Wei-Ting Kuo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Xiaoying Liu
- College of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Fei Wang
- Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
7
|
Sandberg AL, Bond ACS, Bennett LJ, Craig SE, Winski DP, Kirkby LC, Kraemer AR, Kelly KG, Hess ST, Maginnis MS. GPCR Inhibitors Have Antiviral Properties against JC Polyomavirus Infection. Viruses 2024; 16:1559. [PMID: 39459893 PMCID: PMC11512265 DOI: 10.3390/v16101559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/28/2024] Open
Abstract
JC polyomavirus (JCPyV) infects the majority of the population and initially establishes a persistent but asymptomatic infection of the kidneys. In healthy individuals, the infection remains controlled by the host immune system, but for individuals experiencing prolonged immunosuppression, the infection can reactivate and spread to the brain, where it causes progressive multifocal leukoencephalopathy (PML), which is a fatal neurodegenerative disease. Currently, there are no approved therapies to treat PML, and affected individuals suffer rapid motor weakness and cognitive deterioration. To identify novel therapeutic treatments for JCPyV infection, receptor agonists/antagonists identified in a previously published drug screen were evaluated for their antiviral properties. Seven drugs were selected and validated using infectivity assays, and the mechanism of inhibition was further explored for G protein coupled receptor (GPCR)-associated inhibitors due to the role of the GPCR 5-hydroxytryptamine 2 receptors (5-HT2Rs) in JCPyV entry. The inhibitors cetirizine and paroxetine both reduced infection early in the JCPyV infectious cycle. Paroxetine specifically reduced viral internalization through altering the receptor density of 5-HT2CR, inhibiting β-arrestin recruitment to the receptor, and reducing MAPK signaling through ERK. These findings highlight the potential of receptor signaling and viral entry mechanisms as possible targets for antiviral drug development. Further, this research suggests that FDA-approved receptor agonists/antagonists currently used to treat other medical conditions could be repurposed into antivirals for the possible treatment of JCPyV infection and the fatal disease PML.
Collapse
Affiliation(s)
- Amanda L. Sandberg
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Avery C. S. Bond
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Lucas J. Bennett
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Sophie E. Craig
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA;
| | - David P. Winski
- Department of Physics & Astronomy, University of Maine, Orono, ME 04469, USA;
| | - Lara C. Kirkby
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Abby R. Kraemer
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Kristina G. Kelly
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Samuel T. Hess
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA;
- Department of Physics & Astronomy, University of Maine, Orono, ME 04469, USA;
| | - Melissa S. Maginnis
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA;
| |
Collapse
|
8
|
Gurevich VV, Gurevich EV. GPCR-dependent and -independent arrestin signaling. Trends Pharmacol Sci 2024; 45:639-650. [PMID: 38906769 PMCID: PMC11227395 DOI: 10.1016/j.tips.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/23/2024]
Abstract
Biological activity of free arrestins is often overlooked. Based on available data, we compare arrestin-mediated signaling that requires and does not require binding to G-protein-coupled receptors (GPCRs). Receptor-bound arrestins activate ERK1/2, Src, and focal adhesion kinase (FAK). Yet, arrestin-3 regulation of Src family member Fgr does not appear to involve receptors. Free arrestin-3 facilitates the activation of JNK family kinases, preferentially binds E3 ubiquitin ligases Mdm2 and parkin, and facilitates parkin-dependent mitophagy. The binding of arrestins to microtubules and calmodulin and their function in focal adhesion disassembly and apoptosis also do not involve receptors. Biased GPCR ligands and the phosphorylation barcode can only affect receptor-dependent arrestin signaling. Thus, elucidation of receptor dependence or independence of arrestin functions has important scientific and therapeutic implications.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 27232, USA.
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 27232, USA
| |
Collapse
|
9
|
Saito A, Kise R, Inoue A. Generation of Comprehensive GPCR-Transducer-Deficient Cell Lines to Dissect the Complexity of GPCR Signaling. Pharmacol Rev 2024; 76:599-619. [PMID: 38719480 DOI: 10.1124/pharmrev.124.001186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 06/16/2024] Open
Abstract
G-protein-coupled receptors (GPCRs) compose the largest family of transmembrane receptors and are targets of approximately one-third of Food and Drug Administration-approved drugs owing to their involvement in almost all physiologic processes. GPCR signaling occurs through the activation of heterotrimeric G-protein complexes and β-arrestins, both of which serve as transducers, resulting in distinct cellular responses. Despite seeming simple at first glance, accumulating evidence indicates that activation of either transducer is not a straightforward process as a stimulation of a single molecule has the potential to activate multiple signaling branches. The complexity of GPCR signaling arises from the aspects of G-protein-coupling selectivity, biased signaling, interpathway crosstalk, and variable molecular modifications generating these diverse signaling patterns. Numerous questions relative to these aspects of signaling remained unanswered until the recent development of CRISPR genome-editing technology. Such genome editing technology presents opportunities to chronically eliminate the expression of G-protein subunits, β-arrestins, G-protein-coupled receptor kinases (GRKs), and many other signaling nodes in the GPCR pathways at one's convenience. Here, we review the practicality of using CRISPR-derived knockout (KO) cells in the experimental contexts of unraveling the molecular details of GPCR signaling mechanisms. To mention a few, KO cells have revealed the contribution of β-arrestins in ERK activation, Gα protein selectivity, GRK-based regulation of GPCRs, and many more, hence validating its broad applicability in GPCR studies. SIGNIFICANCE STATEMENT: This review emphasizes the practical application of G-protein-coupled receptor (GPCR) transducer knockout (KO) cells in dissecting the intricate regulatory mechanisms of the GPCR signaling network. Currently available cell lines, along with accumulating KO cell lines in diverse cell types, offer valuable resources for systematically elucidating GPCR signaling regulation. Given the association of GPCR signaling with numerous diseases, uncovering the system-based signaling map is crucial for advancing the development of novel drugs targeting specific diseases.
Collapse
Affiliation(s)
- Ayaki Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
10
|
Yanuar R, Semba S, Nezu A, Tanimura A. Muscarinic acetylcholine receptor-mediated phosphorylation of extracellular signal-regulated kinase in HSY salivary ductal cells involves distinct signaling pathways. J Oral Biosci 2024; 66:447-455. [PMID: 38336259 DOI: 10.1016/j.job.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024]
Abstract
OBJECTIVES Typical agonists of G protein-coupled receptors (GPCRs), including muscarinic acetylcholine receptors (mAChRs), activate both G-protein and β-arrestin signaling systems, and are termed balanced agonists. In contrast, biased agonists selectively activate a single pathway, thereby offering therapeutic potential for the specific activation of that pathway. The mAChR agonists carbachol and pilocarpine are known to induce phosphorylation of extracellular signal-regulated kinase-1/2 (ERK1/2) via G-protein-dependent and -independent pathways, respectively. We investigated the involvement of β-arrestin and its downstream mechanisms in the ERK1/2 phosphorylation induced by carbachol and pilocarpine in the human salivary ductal cell line, HSY cells. METHODS HSY cells were stimulated with pilocarpine or carbachol, with or without various inhibitors. The cell lysates were analyzed by western blotting using the antibodies p44/p42MAPK and phosphor-p44/p42MAPK. RESULTS Western blot analysis revealed that carbachol elicited greater stimulation of ERK1/2 phosphorylation compared to pilocarpine. ERK1/2 phosphorylation was inhibited by atropine and gefitinib, suggesting that mAChR activation induces transactivation of epidermal growth factor receptors (EGFR). Moreover, inhibition of carbachol-mediated ERK1/2 phosphorylation was achieved by GF-109203X (a PKC inhibitor), a βARK1/GRK2 inhibitor, barbadin (a β-arrestin inhibitor), pitstop 2 (a clathrin inhibitor), and dynole 34-2 (a dynamin inhibitor). In contrast, pilocarpine-mediated ERK1/2 phosphorylation was only inhibited by barbadin (a β-arrestin inhibitor) and PP2 (a Src inhibitor). CONCLUSION Carbachol activates both G-protein and β-arrestin pathways, whereas pilocarpine exclusively activates the β-arrestin pathway. Additionally, downstream of β-arrestin, carbachol activates clathrin-dependent internalization, while pilocarpine activates Src.
Collapse
Affiliation(s)
- Rezon Yanuar
- Division of Pharmacology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Shingo Semba
- Division of Pharmacology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Akihiro Nezu
- Division of Pharmacology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Akihiko Tanimura
- Division of Pharmacology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan.
| |
Collapse
|
11
|
Guo L, Zhou L, Wei P, Li S, He S, Li D. Emerging Roles of UDP-GalNAc Polypeptide N-Acetylgalactosaminyltransferases in Cardiovascular Disease. Aging Dis 2024; 16:AD.2024.0308. [PMID: 38502587 PMCID: PMC11745429 DOI: 10.14336/ad.2024.0308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
UDP-GalNAc polypeptide N-acetylgalactosaminyltransferases (GalNAc-Ts) catalyze mucin-type O-glycosylation by transferring α-N-acetylgalactosamine (GalNAc) from UDP-GalNAc to Ser or Thr residues of target proteins. This post-translational modification is common in eukaryotes, yet its biological functions remain unclear. Recent studies have identified specific receptors in the heart and vascular wall cells that can be mucin-type O-glycosylated, and there is now substantial evidence confirming that patients with various cardiovascular diseases (CVDs), such as heart failure, coronary artery disease, myocardial hypertrophy, and vascular calcification, exhibit abnormal changes in GalNAc-Ts. This review aims to highlight recent advances in GalNAc-Ts and their roles in the cardiovascular system, intending to provide evidence for clinical treatment and prevention of CVDs.
Collapse
Affiliation(s)
- Liwei Guo
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China.
| | - Lulu Zhou
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Pengcheng Wei
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Shijie Li
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Shanqing He
- Department of Cardiovascular Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Duan Li
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
- Henan Key Biological of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
12
|
Jones RD. Information Transmission in G Protein-Coupled Receptors. Int J Mol Sci 2024; 25:1621. [PMID: 38338905 PMCID: PMC10855935 DOI: 10.3390/ijms25031621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest class of receptors in the human genome and constitute about 30% of all drug targets. In this article, intended for a non-mathematical audience, both experimental observations and new theoretical results are compared in the context of information transmission across the cell membrane. The amount of information actually currently used or projected to be used in clinical settings is a small fraction of the information transmission capacity of the GPCR. This indicates that the number of yet undiscovered drug targets within GPCRs is much larger than what is currently known. Theoretical studies with some experimental validation indicate that localized heat deposition and dissipation are key to the identification of sites and mechanisms for drug action.
Collapse
Affiliation(s)
- Roger D Jones
- European Centre for Living Technology, University of Venice, 30123 Venice, Italy
| |
Collapse
|
13
|
Maharana J, Sano FK, Sarma P, Yadav MK, Duan L, Stepniewski TM, Chaturvedi M, Ranjan A, Singh V, Saha S, Mahajan G, Chami M, Shihoya W, Selent J, Chung KY, Banerjee R, Nureki O, Shukla AK. Molecular insights into atypical modes of β-arrestin interaction with seven transmembrane receptors. Science 2024; 383:101-108. [PMID: 38175886 PMCID: PMC7615931 DOI: 10.1126/science.adj3347] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024]
Abstract
β-arrestins (βarrs) are multifunctional proteins involved in signaling and regulation of seven transmembrane receptors (7TMRs), and their interaction is driven primarily by agonist-induced receptor activation and phosphorylation. Here, we present seven cryo-electron microscopy structures of βarrs either in the basal state, activated by the muscarinic receptor subtype 2 (M2R) through its third intracellular loop, or activated by the βarr-biased decoy D6 receptor (D6R). Combined with biochemical, cellular, and biophysical experiments, these structural snapshots allow the visualization of atypical engagement of βarrs with 7TMRs and also reveal a structural transition in the carboxyl terminus of βarr2 from a β strand to an α helix upon activation by D6R. Our study provides previously unanticipated molecular insights into the structural and functional diversity encoded in 7TMR-βarr complexes with direct implications for exploring novel therapeutic avenues.
Collapse
Affiliation(s)
- Jagannath Maharana
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Fumiya K. Sano
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Parishmita Sarma
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Manish K. Yadav
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Longhan Duan
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Tomasz M. Stepniewski
- Research Program on Biomedical Informatics, Hospital del Mar Research Institute and Pompeu Fabra University, Barcelona, Spain
| | - Madhu Chaturvedi
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Ashutosh Ranjan
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Vinay Singh
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Sayantan Saha
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Gargi Mahajan
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Mohamed Chami
- BioEM Lab, Biozentrum, University of Basel, Basel, Switzerland
| | - Wataru Shihoya
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Jana Selent
- Research Program on Biomedical Informatics, Hospital del Mar Research Institute and Pompeu Fabra University, Barcelona, Spain
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ramanuj Banerjee
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Osamu Nureki
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Arun K. Shukla
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| |
Collapse
|
14
|
Fessl T, Majellaro M, Bondar A. Microscopy and spectroscopy approaches to study GPCR structure and function. Br J Pharmacol 2023. [PMID: 38087925 DOI: 10.1111/bph.16297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/03/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
The GPCR signalling cascade is a key pathway responsible for the signal transduction of a multitude of physical and chemical stimuli, including light, odorants, neurotransmitters and hormones. Understanding the structural and functional properties of the GPCR cascade requires direct observation of signalling processes in high spatial and temporal resolution, with minimal perturbation to endogenous systems. Optical microscopy and spectroscopy techniques are uniquely suited to this purpose because they excel at multiple spatial and temporal scales and can be used in living objects. Here, we review recent developments in microscopy and spectroscopy technologies which enable new insights into GPCR signalling. We focus on advanced techniques with high spatial and temporal resolution, single-molecule methods, labelling strategies and approaches suitable for endogenous systems and large living objects. This review aims to assist researchers in choosing appropriate microscopy and spectroscopy approaches for a variety of applications in the study of cellular signalling.
Collapse
Affiliation(s)
- Tomáš Fessl
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | | | - Alexey Bondar
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
- Laboratory of Microscopy and Histology, Institute of Entomology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| |
Collapse
|
15
|
Steinberg SF. Redox and proteolytic regulation of cardiomyocyte β 1-adrenergic receptors - a novel paradigm for the regulation of catecholamine responsiveness in the heart. Front Immunol 2023; 14:1306467. [PMID: 38111579 PMCID: PMC10726029 DOI: 10.3389/fimmu.2023.1306467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/21/2023] [Indexed: 12/20/2023] Open
Abstract
Conventional models view β1-adrenergic receptors (β1ARs) as full-length proteins that activate signaling pathways that influence contractile function and ventricular remodeling - and are susceptible to agonist-dependent desensitization. This perspective summarizes recent studies from my laboratory showing that post-translational processing of the β1-adrenergic receptor N-terminus results in the accumulation of both full-length and N-terminally truncated forms of the β1AR that differ in their signaling properties. We also implicate oxidative stress and β1AR cleavage by elastase as two novel mechanisms that would (in the setting of cardiac injury or inflammation) lead to altered or decreased β1AR responsiveness.
Collapse
Affiliation(s)
- Susan F. Steinberg
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, United States
| |
Collapse
|
16
|
Tompkins E, Mimic B, Penn RB, Pera T. The biased M3 mAChR ligand PD 102807 mediates qualitatively distinct signaling to regulate airway smooth muscle phenotype. J Biol Chem 2023; 299:105209. [PMID: 37660916 PMCID: PMC10520882 DOI: 10.1016/j.jbc.2023.105209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 08/14/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023] Open
Abstract
Airway smooth muscle (ASM) cells attain a hypercontractile phenotype during obstructive airway diseases. We recently identified a biased M3 muscarinic acetylcholine receptor (mAChR) ligand, PD 102807, that induces GRK-/arrestin-dependent AMP-activated protein kinase (AMPK) activation to inhibit transforming growth factor-β-induced hypercontractile ASM phenotype. Conversely, the balanced mAChR agonist, methacholine (MCh), activates AMPK yet does not regulate ASM phenotype. In the current study, we demonstrate that PD 102807- and MCh-induced AMPK activation both depend on Ca2+/calmodulin-dependent kinase kinases (CaMKKs). However, MCh-induced AMPK activation is calcium-dependent and mediated by CaMKK1 and CaMKK2 isoforms. In contrast, PD 102807-induced signaling is calcium-independent and mediated by the atypical subtype protein kinase C-iota and the CaMKK1 (but not CaMKK2) isoform. Both MCh- and PD 102807-induced AMPK activation involve the AMPK α1 isoform. PD 102807-induced AMPK α1 (but not AMPK α2) isoform activation mediates inhibition of the mammalian target of rapamycin complex 1 (mTORC1) in ASM cells, as demonstrated by increased Raptor (regulatory-associated protein of mTOR) phosphorylation as well as inhibition of phospho-S6 protein and serum response element-luciferase activity. The mTORC1 inhibitor rapamycin and the AMPK activator metformin both mimic the ability of PD 102807 to attenuate transforming growth factor-β-induced α-smooth muscle actin expression (a marker of hypercontractile ASM). These data indicate that PD 102807 transduces a signaling pathway (AMPK-mediated mTORC1 inhibition) qualitatively distinct from canonical M3 mAChR signaling to prevent pathogenic remodeling of ASM, thus demonstrating PD 102807 is a biased M3 mAChR ligand with therapeutic potential for the management of obstructive airway disease.
Collapse
Affiliation(s)
- Eric Tompkins
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania, USA
| | - Bogdana Mimic
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania, USA
| | - Raymond B Penn
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania, USA
| | - Tonio Pera
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
17
|
Jiang Y, Li Y, Fu X, Wu Y, Wang R, Zhao M, Mao C, Shi S. Interplay between G protein-coupled receptors and nanotechnology. Acta Biomater 2023; 169:1-18. [PMID: 37517621 DOI: 10.1016/j.actbio.2023.07.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/15/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
G protein-coupled receptors (GPCRs), as the largest family of membrane receptors, actively modulate plasma membrane and endosomal signalling. Importantly, GPCRs are naturally nanosized, and spontaneously formed nanoaggregates of GPCRs (natural nano-GPCRs) may enhance GPCR-related signalling and functions. Although GPCRs are the molecular targets of the majority of marketed drugs, the poor pharmacokinetics and physicochemical properties of GPCR ligands greatly limit their clinical applicability. Nanotechnology, as versatile techniques, can encapsulate GPCR ligands to assemble synthetic nano-GPCRs to overcome their obstacles, robustly elevating drug efficacy and safety. Moreover, endosomal delivery of GPCR ligands by nanoparticles can precisely initiate sustained endosomal signal transduction, while nanotechnology has been widely utilized for isolation, diagnosis, and detection of GPCRs. In turn, due to overexpression of GPCRs on the surface of various types of cells, GPCR ligands can endow nanoparticles with active targeting capacity for specific cells via ligand-receptor binding and mediate receptor-dependent endocytosis of nanoparticles. This significantly enhances the potency of nanoparticle delivery systems. Therefore, emerging evidence has revealed the interplay between GPCRs and nanoparticles, although investigations into their relationship have been inadequate. This review aims to summarize the interaction between GPCRs and nanotechnology for understanding their mutual influences and utilizing their interplay for biomedical applications. It will provide a fundamental platform for developing powerful and safe GPCR-targeted drugs and nanoparticle systems. STATEMENT OF SIGNIFICANCE: GPCRs as molecular targets for the majority of marketed drugs are naturally nanosized, and even spontaneously form nano aggregations (nano-GPCRs). Nanotechnology has also been applied to construct synthetic nano-GPCRs or detect GPCRs, while endosomal delivery of GPCR ligands by nanoparticles can magnify endosomal signalling. Meanwhile, molecular engineering of nanoparticles with GPCRs or their ligands can modulate membrane binding and endocytosis, powerfully improving the efficacy of nanoparticle system. However, there are rare summaries on the interaction between GPCRs and nanoparticles. This review will not only provide a versatile platform for utilizing nanoparticles to modulate or detect GPCRs, but also facilitate better understanding of the designated value of GPCRs for molecular engineering of biomaterials with GPCRs in therapeutical application.
Collapse
Affiliation(s)
- Yuhong Jiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiujuan Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yue Wu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Rujing Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Canquan Mao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
18
|
Michinaga S, Nagata A, Ogami R, Ogawa Y, Hishinuma S. Differential regulation of histamine H 1 receptor-mediated ERK phosphorylation by G q proteins and arrestins. Biochem Pharmacol 2023; 213:115595. [PMID: 37201878 DOI: 10.1016/j.bcp.2023.115595] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/20/2023]
Abstract
Gq protein-coupled histamine H1 receptors play crucial roles in allergic and inflammatory reactions, in which the phosphorylation of extracellular signal-regulated kinase (ERK) appears to mediate the production of inflammatory cytokines. ERK phosphorylation is regulated by G protein- and arrestin-mediated signal transduction pathways. Here, we aimed to explore how H1 receptor-mediated processes of ERK phosphorylation might be differentially regulated by Gq proteins and arrestins. For this purpose, we evaluated the regulatory mechanism(s) of H1 receptor-mediated ERK phosphorylation in Chinese hamster ovary cells expressing Gq protein- and arrestin-biased mutants of human H1 receptors, S487TR and S487A, in which the Ser487 residue in the C-terminal was truncated and mutated to alanine, respectively. Immunoblotting analysis indicated that histamine-induced ERK phosphorylation was prompt and transient in cells expressing Gq protein-biased S487TR, whereas it was slow and sustained in cells expressing arrestin-biased S487A. Inhibitors of Gq proteins (YM-254890) and protein kinase C (PKC) (GF109203X), and an intracellular Ca2+ chelator (BAPTA-AM) suppressed histamine-induced ERK phosphorylation in cells expressing S487TR, but not those expressing S487A. Conversely, inhibitors of G protein-coupled receptor kinases (GRK2/3) (cmpd101), β-arrestin2 (β-arrestin2 siRNA), clathrin (hypertonic sucrose), Raf (LY3009120), and MEK (U0126) suppressed histamine-induced ERK phosphorylation in cells expressing S487A, but not those expressing S487TR. These results suggest that H1 receptor-mediated ERK phosphorylation might be differentially regulated by the Gq protein/Ca2+/PKC and GRK/arrestin/clathrin/Raf/MEK pathways to potentially determine the early and late phases of histamine-induced allergic and inflammatory responses, respectively.
Collapse
Affiliation(s)
- Shotaro Michinaga
- Department of Pharmacodynamics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan
| | - Ayaka Nagata
- Department of Pharmacodynamics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan
| | - Ryosuke Ogami
- Department of Pharmacodynamics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan
| | - Yasuhiro Ogawa
- Department of Pharmacodynamics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan
| | - Shigeru Hishinuma
- Department of Pharmacodynamics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan.
| |
Collapse
|
19
|
Fouillen A, Bous J, Granier S, Mouillac B, Sounier R. Bringing GPCR Structural Biology to Medical Applications: Insights from Both V2 Vasopressin and Mu-Opioid Receptors. MEMBRANES 2023; 13:606. [PMID: 37367810 PMCID: PMC10303988 DOI: 10.3390/membranes13060606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023]
Abstract
G-protein coupled receptors (GPCRs) are versatile signaling proteins that regulate key physiological processes in response to a wide variety of extracellular stimuli. The last decade has seen a revolution in the structural biology of clinically important GPCRs. Indeed, the improvement in molecular and biochemical methods to study GPCRs and their transducer complexes, together with advances in cryo-electron microscopy, NMR development, and progress in molecular dynamic simulations, have led to a better understanding of their regulation by ligands of different efficacy and bias. This has also renewed a great interest in GPCR drug discovery, such as finding biased ligands that can either promote or not promote specific regulations. In this review, we focus on two therapeutically relevant GPCR targets, the V2 vasopressin receptor (V2R) and the mu-opioid receptor (µOR), to shed light on the recent structural biology studies and show the impact of this integrative approach on the determination of new potential clinical effective compounds.
Collapse
Affiliation(s)
- Aurélien Fouillen
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34000 Montpellier, France; (A.F.); (S.G.); (B.M.)
- Centre de Biochimie Structurale (CBS), Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France
| | - Julien Bous
- Section of Receptor Biology & Signaling, Department of Physiology & Pharmacology, Karolinska Institutet, 17165 Stockholm, Sweden;
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34000 Montpellier, France; (A.F.); (S.G.); (B.M.)
| | - Bernard Mouillac
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34000 Montpellier, France; (A.F.); (S.G.); (B.M.)
| | - Remy Sounier
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34000 Montpellier, France; (A.F.); (S.G.); (B.M.)
| |
Collapse
|
20
|
Chen G, Obal D. Detecting and measuring of GPCR signaling - comparison of human induced pluripotent stem cells and immortal cell lines. Front Endocrinol (Lausanne) 2023; 14:1179600. [PMID: 37293485 PMCID: PMC10244570 DOI: 10.3389/fendo.2023.1179600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/12/2023] [Indexed: 06/10/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family of transmembrane proteins that play a major role in many physiological processes, and thus GPCR-targeted drug development has been widely promoted. Although research findings generated in immortal cell lines have contributed to the advancement of the GPCR field, the homogenous genetic backgrounds, and the overexpression of GPCRs in these cell lines make it difficult to correlate the results with clinical patients. Human induced pluripotent stem cells (hiPSCs) have the potential to overcome these limitations, because they contain patient specific genetic information and can differentiate into numerous cell types. To detect GPCRs in hiPSCs, highly selective labeling and sensitive imaging techniques are required. This review summarizes existing resonance energy transfer and protein complementation assay technologies, as well as existing and new labeling methods. The difficulties of extending existing detection methods to hiPSCs are discussed, as well as the potential of hiPSCs to expand GPCR research towards personalized medicine.
Collapse
Affiliation(s)
- Gaoxian Chen
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Detlef Obal
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| |
Collapse
|
21
|
Wong TS, Li G, Li S, Gao W, Chen G, Gan S, Zhang M, Li H, Wu S, Du Y. G protein-coupled receptors in neurodegenerative diseases and psychiatric disorders. Signal Transduct Target Ther 2023; 8:177. [PMID: 37137892 PMCID: PMC10154768 DOI: 10.1038/s41392-023-01427-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Neuropsychiatric disorders are multifactorial disorders with diverse aetiological factors. Identifying treatment targets is challenging because the diseases are resulting from heterogeneous biological, genetic, and environmental factors. Nevertheless, the increasing understanding of G protein-coupled receptor (GPCR) opens a new possibility in drug discovery. Harnessing our knowledge of molecular mechanisms and structural information of GPCRs will be advantageous for developing effective drugs. This review provides an overview of the role of GPCRs in various neurodegenerative and psychiatric diseases. Besides, we highlight the emerging opportunities of novel GPCR targets and address recent progress in GPCR drug development.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Wei Gao
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Shiyi Gan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Manzhan Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China.
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China.
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, 518116, Shenzhen, Guangdong, China.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China.
| |
Collapse
|
22
|
Martinez JM, Shen A, Xu B, Jovanovic A, de Chabot J, Zhang J, Xiang YK. Arrestin-dependent nuclear export of phosphodiesterase 4D promotes GPCR-induced nuclear cAMP signaling required for learning and memory. Sci Signal 2023; 16:eade3380. [PMID: 36976866 PMCID: PMC10404024 DOI: 10.1126/scisignal.ade3380] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/07/2023] [Indexed: 03/30/2023]
Abstract
G protein-coupled receptors (GPCRs) promote the expression of immediate early genes required for learning and memory. Here, we showed that β2-adrenergic receptor (β2AR) stimulation induced the nuclear export of phosphodiesterase 4D5 (PDE4D5), an enzyme that degrades the second messenger cAMP, to enable memory consolidation. We demonstrated that the endocytosis of β2AR phosphorylated by GPCR kinases (GRKs) mediated arrestin3-dependent nuclear export of PDE4D5, which was critical for promoting nuclear cAMP signaling and gene expression in hippocampal neurons for memory consolidation. Inhibition of the arrestin3-PDE4D5 association prevented β2AR-induced nuclear cAMP signaling without affecting receptor endocytosis. Direct PDE4 inhibition rescued β2AR-induced nuclear cAMP signaling and ameliorated memory deficits in mice expressing a form of the β2AR that could not be phosphorylated by GRKs. These data reveal how β2AR phosphorylated by endosomal GRK promotes the nuclear export of PDE4D5, leading to nuclear cAMP signaling, changes in gene expression, and memory consolidation. This study also highlights the translocation of PDEs as a mechanism to promote cAMP signaling in specific subcellular locations downstream of GPCR activation.
Collapse
Affiliation(s)
- Joseph M. Martinez
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Ao Shen
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
- School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Bing Xu
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
- VA Northern California Health Care System, Mather, CA, 95655, USA
| | - Aleksandra Jovanovic
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Josephine de Chabot
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Jin Zhang
- Department of Pharmacology, University of California at San Diego, San Diego, CA, 92093, USA
| | - Yang K. Xiang
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
- VA Northern California Health Care System, Mather, CA, 95655, USA
| |
Collapse
|
23
|
Matarrese P, Maccari S, Gambardella L, Vona R, Barbagallo F, Vezzi V, Stati T, Grò MC, Giovannetti A, Catalano L, Molinari P, Marano G, Ambrosio C. Benzodiazepine diazepam regulates cell surface β1-adrenergic receptor density in human monocytes. Eur J Pharmacol 2023; 948:175700. [PMID: 37001579 DOI: 10.1016/j.ejphar.2023.175700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
Downregulation of cell surface β-adrenergic receptors (β-AR) is an important adaptive response that prevents deleterious effects of receptor overstimulation. Various factors including reactive oxygen species cause β-AR downregulation. In this study, we evaluated the effects of ligands of the peripheral benzodiazepine receptor (PBR), a key protein in regulating oxidative stress, on surface density of endogenous β1-and β2-ARs in highly differentiated cells such as human monocytes, which express both β-AR subtypes. β-AR expression in human monocytes was evaluated by flow cytometry, qPCR and western blotting. Monocyte treatment with β-AR agonist isoproterenol did not change surface β1-AR density while downregulating surface β2-AR density. This effect was antagonized by the β-blocker propranolol. An opposite response was observed with benzodiazepine diazepam that led to a time-dependent reduction in β1-AR density. In particular, while no significant downregulation was observed after 3 h of treatment, only 63% of β1-ARs were still present on the cell surface after 48 h of treatment with diazepam at 1 μM. Treatment with the PBR antagonist PK11195, but not with propranolol, antagonized the effects of diazepam. No change in β1-AR-mRNA or protein levels was observed at any time after diazepam treatment. We also found that diazepam did not affect Gs-protein or β-arrestin-2 recruitment for both β-ARs in engineered fibroblasts, further suggesting that diazepam activity on β1-AR density is mediated by PBR. Finally, no sex-related differences were found. Collectively, these results indicate that monocyte β1-ARs are resistant to catecholamine-mediated downregulation and suggest that PBR plays an important role in regulating β1-AR density.
Collapse
|
24
|
Cabral-Dias R, Antonescu CN. Control of phosphatidylinositol-3-kinase signaling by nanoscale membrane compartmentalization. Bioessays 2023; 45:e2200196. [PMID: 36567275 DOI: 10.1002/bies.202200196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 09/12/2022] [Accepted: 12/13/2022] [Indexed: 12/27/2022]
Abstract
Phosphatidylinositol-3-kinases (PI3Ks) are lipid kinases that produce 3-phosphorylated derivatives of phosphatidylinositol upon activation by various cues. These 3-phosphorylated lipids bind to various protein effectors to control many cellular functions. Lipid phosphatases such as phosphatase and tensin homolog (PTEN) terminate PI3K-derived signals and are critical to ensure appropriate signaling outcomes. Many lines of evidence indicate that PI3Ks and PTEN, as well as some specific lipid effectors are highly compartmentalized, either in plasma membrane nanodomains or in endosomal compartments. We examine the evidence for specific recruitment of PI3Ks, PTEN, and other related enzymes to membrane nanodomains and endocytic compartments. We then examine the hypothesis that scaffolding of the sources (PI3Ks), terminators (PTEN), and effectors of these lipid signals with a common plasma membrane nanodomain may achieve highly localized lipid signaling and ensure selective activation of specific effectors. This highlights the importance of spatial regulation of PI3K signaling in various physiological and disease contexts.
Collapse
Affiliation(s)
- Rebecca Cabral-Dias
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Cui Y, Zhang X, Li X, Lin J. Multiscale microscopy to decipher plant cell structure and dynamics. THE NEW PHYTOLOGIST 2023; 237:1980-1997. [PMID: 36477856 DOI: 10.1111/nph.18641] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 11/09/2022] [Indexed: 06/17/2023]
Abstract
New imaging methodologies with high contrast and molecular specificity allow researchers to analyze dynamic processes in plant cells at multiple scales, from single protein and RNA molecules to organelles and cells, to whole organs and tissues. These techniques produce informative images and quantitative data on molecular dynamics to address questions that cannot be answered by conventional biochemical assays. Here, we review selected microscopy techniques, focusing on their basic principles and applications in plant science, discussing the pros and cons of each technique, and introducing methods for quantitative analysis. This review thus provides guidance for plant scientists in selecting the most appropriate techniques to decipher structures and dynamic processes at different levels, from protein dynamics to morphogenesis.
Collapse
Affiliation(s)
- Yaning Cui
- National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing, 100083, China
- College of Biological Sciences & Biotechnology, Beijing Forestry University, Beijing, 100083, China
| | - Xi Zhang
- National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing, 100083, China
- College of Biological Sciences & Biotechnology, Beijing Forestry University, Beijing, 100083, China
| | - Xiaojuan Li
- National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing, 100083, China
- College of Biological Sciences & Biotechnology, Beijing Forestry University, Beijing, 100083, China
| | - Jinxing Lin
- National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing, 100083, China
- College of Biological Sciences & Biotechnology, Beijing Forestry University, Beijing, 100083, China
| |
Collapse
|
26
|
Kurihara Y, Ekimoto T, Gordon CT, Uchijima Y, Sugiyama R, Kitazawa T, Iwase A, Kotani R, Asai R, Pingault V, Ikeguchi M, Amiel J, Kurihara H. Mandibulofacial dysostosis with alopecia results from ETAR gain-of-function mutations via allosteric effects on ligand binding. J Clin Invest 2023; 133:151536. [PMID: 36637912 PMCID: PMC9927936 DOI: 10.1172/jci151536] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 12/16/2022] [Indexed: 01/14/2023] Open
Abstract
Mutations of G protein-coupled receptors (GPCRs) cause various human diseases, but the mechanistic details are limited. Here, we establish p.E303K in the gene encoding the endothelin receptor type A (ETAR/EDNRA) as a recurrent mutation causing mandibulofacial dysostosis with alopecia (MFDA), with craniofacial changes similar to those caused by p.Y129F. Mouse models carrying either of these missense mutations exhibited a partial maxillary-to-mandibular transformation, which was rescued by deleting the ligand endothelin 3 (ET3/EDN3). Pharmacological experiments confirmed the causative ETAR mutations as gain of function, dependent on ET3. To elucidate how an amino acid substitution far from the ligand binding site can increase ligand affinity, we used molecular dynamics (MD) simulations. E303 is located at the intracellular end of transmembrane domain 6, and its replacement by a lysine increased flexibility of this portion of the helix, thus favoring G protein binding and leading to G protein-mediated enhancement of agonist affinity. The Y129F mutation located under the ligand binding pocket reduced the sodium-water network, thereby affecting the extracellular portion of helices in favor of ET3 binding. These findings provide insight into the pathogenesis of MFDA and into allosteric mechanisms regulating GPCR function, which may provide the basis for drug design targeting GPCRs.
Collapse
Affiliation(s)
- Yukiko Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toru Ekimoto
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | | | - Yasunobu Uchijima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryo Sugiyama
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taro Kitazawa
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akiyasu Iwase
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Risa Kotani
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Medical Science, Graduate School of Medicine, University of Hiroshima, Hiroshima, Japan
| | - Rieko Asai
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Véronique Pingault
- Department of Genomic Medicine for Rare Diseases, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Mitsunori Ikeguchi
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.,Center for Computational Science, RIKEN, Yokohama, Japan
| | - Jeanne Amiel
- INSERM UMR 1163, Institut Imagine and Université Paris-Cité, Paris, France.,Department of Genomic Medicine for Rare Diseases, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
27
|
Shiraki A, Shimizu S. The molecular associations in clathrin-coated pit regulate β-arrestin-mediated MAPK signaling downstream of μ-opioid receptor. Biochem Biophys Res Commun 2023; 640:64-72. [PMID: 36502633 DOI: 10.1016/j.bbrc.2022.11.098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
It has been thought that μ-opioid receptors (MOPs) activate the G protein-mediated analgesic pathway and β-arrestin 2-mediated side effect pathway; however, ligands that only minimally recruit β-arrestin 2 to MOPs may also cause opioid side effects. Moreover, such side effects have been induced in mutant mice lacking β-arrestin 2 or expressing phosphorylation-deficient MOPs that do not recruit β-arrestin 2. These findings raise the critical question of whether β-arrestin 2 recruitment to MOP triggers side effects. Here, we show that β-arrestin 1 and 2 are essential in the efficient activation of the Gi/o-mediated MAPK signaling at MOP. Moreover, the magnitude of β-arrestin-mediated signals is not correlated with the magnitude of phosphorylation of the carboxyl-terminal of MOP, which is used to evaluate the β-arrestin bias of a ligand. Instead, the molecular association with β2-adaptin and clathrin heavy chain in the formation of clathrin-coated pits is essential for β-arrestin to activate MAPK signaling. Our findings provide insights into G protein-coupled receptor-mediated signaling and further highlight a concept that the accumulation of molecules required for endocytosis is critical for activating intracellular signaling.
Collapse
Affiliation(s)
- Atsuko Shiraki
- Department of Anesthesia, Kyoto University Hospital, Kyoto City, Japan
| | - Satoshi Shimizu
- Department of Anesthesia, Kyoto University Hospital, Kyoto City, Japan.
| |
Collapse
|
28
|
Dos Santos Claro PA, Silbermins M, Inda C, Silberstein S. CRHR1 endocytosis: Spatiotemporal regulation of receptor signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:229-260. [PMID: 36813360 DOI: 10.1016/bs.pmbts.2022.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Corticotropin releasing hormone (CRH) is crucial for basal and stress-initiated reactions in the hypothalamic-pituitary-adrenal axis (HPA) and extrahypothalamic brain circuits, where it acts as a neuromodulator to organize behavioral and humoral responses to stress. We review and describe cellular components and molecular mechanisms involved in CRH system signaling through G protein-coupled receptors (GPCRs) CRHR1 and CRHR2, under the current view of GPCR signaling from the plasma membrane but also from intracellular compartments, which establish the bases of signal resolution in space and time. Focus is placed on latest studies of CRHR1 signaling in physiologically significant contexts of the neurohormone function that disclosed new mechanistic features of cAMP production and ERK1/2 activation. We also introduce in a brief overview the pathophysiological function of the CRH system, underlining the need for a complete characterization of CRHRs signaling to design new and specific therapies for stress-related disorders.
Collapse
Affiliation(s)
- Paula A Dos Santos Claro
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Micaela Silbermins
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carolina Inda
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina; Octamer SRL, Buenos Aires, Argentina
| | - Susana Silberstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
29
|
Bruna-Gauchoux J, Montagnac G. Constraints and frustration in the clathrin-dependent endocytosis pathway. C R Biol 2022; 345:43-56. [PMID: 36847464 DOI: 10.5802/crbiol.88] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022]
Abstract
Clathrin-dependent endocytosis is the major pathway for the entry of most surface receptors and their ligands. It is controlled by clathrin-coated structures that are endowed with the ability to cluster receptors and locally bend the plasma membrane, leading to the formation of receptor-containing vesicles budding into the cytoplasm. This canonical role of clathrin-coated structures has been repeatedly demonstrated to play a fundamental role in a wide range of aspects of cell physiology. However, it is now clearly established that the ability of clathrin-coated structures to bend the membrane can be disrupted. In addition to chemical or genetic alterations, many environmental conditions can physically prevent or slow membrane deformation and/or budding of clathrin-coated structures. The resulting frustrated endocytosis is not only a passive consequence but serves very specific and important cellular functions. Here we provide a historical perspective as well as a definition of frustrated endocytosis in the clathrin pathway before describing its causes and many functional consequences.
Collapse
|
30
|
Janetzko J, Kise R, Barsi-Rhyne B, Siepe DH, Heydenreich FM, Kawakami K, Masureel M, Maeda S, Garcia KC, von Zastrow M, Inoue A, Kobilka BK. Membrane phosphoinositides regulate GPCR-β-arrestin complex assembly and dynamics. Cell 2022; 185:4560-4573.e19. [PMID: 36368322 PMCID: PMC10030194 DOI: 10.1016/j.cell.2022.10.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/22/2022] [Accepted: 10/14/2022] [Indexed: 11/11/2022]
Abstract
Binding of arrestin to phosphorylated G protein-coupled receptors (GPCRs) is crucial for modulating signaling. Once internalized, some GPCRs remain complexed with β-arrestins, while others interact only transiently; this difference affects GPCR signaling and recycling. Cell-based and in vitro biophysical assays reveal the role of membrane phosphoinositides (PIPs) in β-arrestin recruitment and GPCR-β-arrestin complex dynamics. We find that GPCRs broadly stratify into two groups, one that requires PIP binding for β-arrestin recruitment and one that does not. Plasma membrane PIPs potentiate an active conformation of β-arrestin and stabilize GPCR-β-arrestin complexes by promoting a fully engaged state of the complex. As allosteric modulators of GPCR-β-arrestin complex dynamics, membrane PIPs allow for additional conformational diversity beyond that imposed by GPCR phosphorylation alone. For GPCRs that require membrane PIP binding for β-arrestin recruitment, this provides a mechanism for β-arrestin release upon translocation of the GPCR to endosomes, allowing for its rapid recycling.
Collapse
Affiliation(s)
- John Janetzko
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Benjamin Barsi-Rhyne
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, School of Medicine, San Francisco, CA 94158, USA; Department of Psychiatry, University of California, San Francisco, School of Medicine, San Francisco, CA 94158, USA
| | - Dirk H Siepe
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Franziska M Heydenreich
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Matthieu Masureel
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shoji Maeda
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark von Zastrow
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, School of Medicine, San Francisco, CA 94158, USA; Department of Psychiatry, University of California, San Francisco, School of Medicine, San Francisco, CA 94158, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
31
|
Eiger DS, Boldizsar N, Honeycutt CC, Gardner J, Kirchner S, Hicks C, Choi I, Pham U, Zheng K, Warman A, Smith JS, Zhang JY, Rajagopal S. Location bias contributes to functionally selective responses of biased CXCR3 agonists. Nat Commun 2022; 13:5846. [PMID: 36195635 PMCID: PMC9532441 DOI: 10.1038/s41467-022-33569-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 09/13/2022] [Indexed: 01/19/2023] Open
Abstract
Some G protein-coupled receptor (GPCR) ligands act as "biased agonists" that preferentially activate specific signaling transducers over others. Although GPCRs are primarily found at the plasma membrane, GPCRs can traffic to and signal from many subcellular compartments. Here, we determine that differential subcellular signaling contributes to the biased signaling generated by three endogenous ligands of the GPCR CXC chemokine receptor 3 (CXCR3). The signaling profile of CXCR3 changes as it traffics from the plasma membrane to endosomes in a ligand-specific manner. Endosomal signaling is critical for biased activation of G proteins, β-arrestins, and extracellular-signal-regulated kinase (ERK). In CD8 + T cells, the chemokines promote unique transcriptional responses predicted to regulate inflammatory pathways. In a mouse model of contact hypersensitivity, β-arrestin-biased CXCR3-mediated inflammation is dependent on receptor internalization. Our work demonstrates that differential subcellular signaling is critical to the overall biased response observed at CXCR3, which has important implications for drugs targeting chemokine receptors and other GPCRs.
Collapse
Affiliation(s)
| | | | | | - Julia Gardner
- Trinity College, Duke University, Durham, NC, 27710, USA
| | - Stephen Kirchner
- Department of Dermatology, Duke University, Durham, NC, 27707, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, 27707, USA
| | - Chloe Hicks
- Trinity College, Duke University, Durham, NC, 27710, USA
| | - Issac Choi
- Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Uyen Pham
- Department of Biochemistry, Duke University, Durham, NC, 27710, USA
| | - Kevin Zheng
- Harvard Medical School, Boston, MA, 02115, USA
| | - Anmol Warman
- Trinity College, Duke University, Durham, NC, 27710, USA
| | - Jeffrey S Smith
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Dermatology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Dermatology Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Dermatology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Jennifer Y Zhang
- Department of Dermatology, Duke University, Durham, NC, 27707, USA
- Department of Pathology, Duke University, Durham, NC, 27710, USA
| | - Sudarshan Rajagopal
- Department of Biochemistry, Duke University, Durham, NC, 27710, USA.
- Department of Medicine, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
32
|
Bous J, Fouillen A, Orcel H, Trapani S, Cong X, Fontanel S, Saint-Paul J, Lai-Kee-Him J, Urbach S, Sibille N, Sounier R, Granier S, Mouillac B, Bron P. Structure of the vasopressin hormone-V2 receptor-β-arrestin1 ternary complex. SCIENCE ADVANCES 2022; 8:eabo7761. [PMID: 36054364 PMCID: PMC10866553 DOI: 10.1126/sciadv.abo7761] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Arrestins interact with G protein-coupled receptors (GPCRs) to stop G protein activation and to initiate key signaling pathways. Recent structural studies shed light on the molecular mechanisms involved in GPCR-arrestin coupling, but whether this process is conserved among GPCRs is poorly understood. Here, we report the cryo-electron microscopy active structure of the wild-type arginine-vasopressin V2 receptor (V2R) in complex with β-arrestin1. It reveals an atypical position of β-arrestin1 compared to previously described GPCR-arrestin assemblies, associated with an original V2R/β-arrestin1 interface involving all receptor intracellular loops. Phosphorylated sites of the V2R carboxyl terminus are clearly identified and interact extensively with the β-arrestin1 N-lobe, in agreement with structural data obtained with chimeric or synthetic systems. Overall, these findings highlight a notable structural variability among GPCR-arrestin signaling complexes.
Collapse
Affiliation(s)
- Julien Bous
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Aurélien Fouillen
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Hélène Orcel
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Stefano Trapani
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Xiaojing Cong
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Simon Fontanel
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Julie Saint-Paul
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Joséphine Lai-Kee-Him
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Serge Urbach
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Nathalie Sibille
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Rémy Sounier
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Bernard Mouillac
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Patrick Bron
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
33
|
Steinberg SF. N-Tertaining a New Signaling Paradigm for the Cardiomyocyte β 1 -Adrenergic Receptor. J Cardiovasc Pharmacol 2022; 80:328-333. [PMID: 35099166 PMCID: PMC9170829 DOI: 10.1097/fjc.0000000000001194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/20/2021] [Indexed: 01/31/2023]
Abstract
ABSTRACT β 1 -adrenergic receptors (β 1 ARs) are the principle mediators of catecholamine actions in cardiomyocytes. β 1 ARs rapidly adjust cardiac output and provide short-term hemodynamic support for the failing heart by activating a Gs-adenylyl cyclase pathway that increases 3'-5'-cyclic adenosine monophosphate and leads to the activation of protein kinase A and the phosphorylation of substrates involved in excitation-contraction coupling. However, chronic persistent β 1 AR activation in the setting of heart failure leads to a spectrum of maladaptive changes that contribute to the evolution of heart failure. The molecular basis for β 1 AR-driven maladaptive responses remains uncertain because chronic persistent β 1 AR activation has been linked to the activation of both proapoptotic and antiapoptotic signaling pathways. Of note, studies to date have been predicated on the assumption that β 1 ARs signal exclusively as full-length receptor proteins. Our recent studies show that β 1 ARs are detected as both full-length and N-terminally truncated species in cardiomyocytes, that N-terminal cleavage is regulated by O-glycan modifications at specific sites on the β 1 AR N-terminus, and that N-terminally truncated β 1 ARs remain signaling competent, but their signaling properties differ from those of the full-length β 1 AR. The N-terminally truncated form of the β 1 AR constitutively activates the protein kinase B signaling pathway and confers protection against doxorubicin-dependent apoptosis in cardiomyocytes. These studies identify a novel signaling paradigm for the β 1 AR, implicating the N-terminus as a heretofore-unrecognized structural determinant of β 1 AR responsiveness that could be pharmacologically targeted for therapeutic advantage.
Collapse
|
34
|
Baidya M, Chaturvedi M, Dwivedi-Agnihotri H, Ranjan A, Devost D, Namkung Y, Stepniewski TM, Pandey S, Baruah M, Panigrahi B, Sarma P, Yadav MK, Maharana J, Banerjee R, Kawakami K, Inoue A, Selent J, Laporte SA, Hébert TE, Shukla AK. Allosteric modulation of GPCR-induced β-arrestin trafficking and signaling by a synthetic intrabody. Nat Commun 2022; 13:4634. [PMID: 35941121 PMCID: PMC9360436 DOI: 10.1038/s41467-022-32386-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 07/28/2022] [Indexed: 12/28/2022] Open
Abstract
Agonist-induced phosphorylation of G protein-coupled receptors (GPCRs) is a primary determinant of β-arrestin (βarr) recruitment and trafficking. For several GPCRs such as the vasopressin receptor subtype 2 (V2R), agonist-stimulation first drives the translocation of βarrs to the plasma membrane, followed by endosomal trafficking, which is generally considered to be orchestrated by multiple phosphorylation sites. We have previously shown that mutation of a single phosphorylation site in the V2R (i.e., V2RT360A) results in near-complete loss of βarr translocation to endosomes despite robust recruitment to the plasma membrane, and compromised ERK1/2 activation. Here, we discover that a synthetic intrabody (Ib30), which selectively recognizes activated βarr1, efficiently rescues the endosomal trafficking of βarr1 and ERK1/2 activation for V2RT360A. Molecular dynamics simulations reveal that Ib30 enriches active-like βarr1 conformation with respect to the inter-domain rotation, and cellular assays demonstrate that it also enhances βarr1-β2-adaptin interaction. Our data provide an experimental framework to positively modulate the receptor-transducer-effector axis for GPCRs using intrabodies, which can be potentially integrated in the paradigm of GPCR-targeted drug discovery.
Collapse
Affiliation(s)
- Mithu Baidya
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Madhu Chaturvedi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Hemlata Dwivedi-Agnihotri
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Ashutosh Ranjan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Dominic Devost
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Yoon Namkung
- Department of Medicine, McGill University Health Center, McGill University, Montréal, QC, H4A 3J1, Canada
| | - Tomasz Maciej Stepniewski
- Research Program on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences of Pompeu Fabra University (UPF)-Hospital del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Shubhi Pandey
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Minakshi Baruah
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Bhanupriya Panigrahi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Parishmita Sarma
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Manish K Yadav
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Jagannath Maharana
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Ramanuj Banerjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Jana Selent
- Research Program on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences of Pompeu Fabra University (UPF)-Hospital del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain
| | - Stéphane A Laporte
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, H3G 1Y6, Canada
- Department of Medicine, McGill University Health Center, McGill University, Montréal, QC, H4A 3J1, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India.
| |
Collapse
|
35
|
Emerging structural insights into GPCR-β-arrestin interaction and functional outcomes. Curr Opin Struct Biol 2022; 75:102406. [PMID: 35738165 PMCID: PMC7614528 DOI: 10.1016/j.sbi.2022.102406] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/28/2022] [Accepted: 05/19/2022] [Indexed: 11/22/2022]
Abstract
Agonist-induced recruitment of β-arrestins (βarrs) to G protein-coupled receptors (GPCRs) plays a central role in regulating the spatio-temporal aspects of GPCR signaling. Several recent studies have provided novel structural and functional insights into our understanding of GPCR-βarr interaction, subsequent βarr activation and resulting functional outcomes. In this review, we discuss these recent advances with a particular emphasis on recognition of receptor-bound phosphates by βarrs, the emerging concept of spatial positioning of key phosphorylation sites, the conformational transition in βarrs during partial to full-engagement, and structural differences driving functional outcomes of βarr isoforms. We also highlight the key directions that require further investigation going forward to fully understand the structural mechanisms driving βarr activation and functional responses.
Collapse
|
36
|
Kim GT, Kim EY, Shin SH, Lee H, Lee SH, Sohn KY, Kim JW. Suppression of tumor progression by thioredoxin-interacting protein-dependent adenosine 2B receptor degradation in a PLAG-treated Lewis lung carcinoma-1 model of non-small cell lung cancer. Neoplasia 2022; 31:100815. [PMID: 35728512 PMCID: PMC9209866 DOI: 10.1016/j.neo.2022.100815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 05/23/2022] [Accepted: 06/08/2022] [Indexed: 11/03/2022]
Abstract
PLAG effectively inhibited excessive growth of LLC1 cells in an NSCLC model. PLAG inhibited tumor growth by inducing adenosine 2B receptor (A2BR) degradation. Unlike antagonists, PLAG terminates rather than suppresses signaling pathways. A2BR degradation by PLAG occurs through expression and re-localization of TXNIP.
Extracellular adenosine in the tumor microenvironment plays a vital role in cancer development. Specifically, activation of adenosine receptors affects tumor cell growth and adenosine release. We examined the anti-tumor efficacy of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) in animal models, revealing the role of PLAG in inhibiting tumor progression by promoting the degradation of adenosine 2B receptors (A2BRs) in tumors. PLAG induced the expression of thioredoxin-interacting protein (TXNIP), a type of α-arrestin that accelerates A2BR internalization by interacting with A2BR complexes containing β-arrestin. Engulfed receptors bound to TXNIP were rapidly degraded after E3 ligase recruitment and ubiquitination, resulting in early termination of intracellular signals that promote tumor overgrowth. However, in control cancer cells, A2BRs bound to protein phosphatase 2A and were returned to the cell membrane instead of being degraded, resulting in continuous receptor-mediated signaling by pathways including the Raf-Erk axis, which promotes tumor proliferation. A TXNIP-silenced cell-implanted mouse model and TXNIP knockout (KO) mice were used to verify that PLAG-mediated suppression of tumor progression is dependent on TXNIP expression. Increased tumor growth was observed in TXNIP-silenced cell-implanted mice, and the anti-tumor effects of PLAG, including delayed tumor overgrowth, were greatly reduced. However, the anti-tumor effects of PLAG were observed in cancer cell-implanted TXNIP-KO mice, which indicates that PLAG produces anti-tumor effects by enhancing TXNIP expression in tumor cells. These essential functions of PLAG, including delaying tumor growth via A2BR degradation, suggest innovative directions for anticancer drug development.
Collapse
Affiliation(s)
- Guen Tae Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea.
| |
Collapse
|
37
|
Tang X, Bian J, Li Z. Post-Translational Modifications in GPCR Internalization. Am J Physiol Cell Physiol 2022; 323:C84-C94. [PMID: 35613355 DOI: 10.1152/ajpcell.00015.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of membrane receptors that serve as the most important drug targets. Classically, GPCR internalization has been considered to lead to receptor desensitization. However, many studies over the past decade have reported that internalized membrane receptors can trigger distinct signal activation. The "internalized activation" provides a completely new understanding for the receptor internalization, the mechanism of physiology/pathology and novel drug targets for precision medicine. GPCR internalization undergoes a series of strict regulations, especially by post-translational modifications (PTMs). Here, this review summarizes different PTMs in GPCR internalization and analyzes their significance in GPCR internalization dynamics, internalization routes, post-internalization fates and related diseases, which will offer new insights into the regulatory mechanism of GPCR signaling and novel drug targets for precision medicine.
Collapse
Affiliation(s)
- Xueqing Tang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Jingwei Bian
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Zijian Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.,Department of Pharmacy, Peking University Third Hospital, Beijing, China
| |
Collapse
|
38
|
Thor D. G protein-coupled receptors as regulators of pancreatic islet functionality. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119235. [PMID: 35151663 DOI: 10.1016/j.bbamcr.2022.119235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 01/03/2023]
Abstract
Glucose homeostasis is maintained by hormones secreted from different types of pancreatic islets and its dysregulation can result in diseases including diabetes mellitus. The secretion of hormones from pancreatic islets is highly complex and tightly controlled by G protein-coupled receptors (GPCRs). Moreover, GPCR signaling may play a role in enhancing islet cell replication and proliferation. Thus, targeting GPCRs offers a promising strategy for regulating the functionality of pancreatic islets. Here, available RNAseq datasets from human and mouse islets were used to identify the GPCR expression profile and the impact of GPCR signaling for normal islet functionality is discussed.
Collapse
Affiliation(s)
- Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany.
| |
Collapse
|
39
|
Cabral-Dias R, Lucarelli S, Zak K, Rahmani S, Judge G, Abousawan J, DiGiovanni LF, Vural D, Anderson KE, Sugiyama MG, Genc G, Hong W, Botelho RJ, Fairn GD, Kim PK, Antonescu CN. Fyn and TOM1L1 are recruited to clathrin-coated pits and regulate Akt signaling. J Cell Biol 2022; 221:213045. [PMID: 35238864 PMCID: PMC8899389 DOI: 10.1083/jcb.201808181] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 12/24/2022] Open
Abstract
The epidermal growth factor (EGF) receptor (EGFR) controls many aspects of cell physiology. EGF binding to EGFR elicits the membrane recruitment and activation of phosphatidylinositol-3-kinase, leading to Akt phosphorylation and activation. Concomitantly, EGFR is recruited to clathrin-coated pits (CCPs), eventually leading to receptor endocytosis. Previous work uncovered that clathrin, but not receptor endocytosis, is required for EGF-stimulated Akt activation, and that some EGFR signals are enriched in CCPs. Here, we examine how CCPs control EGFR signaling. The signaling adaptor TOM1L1 and the Src-family kinase Fyn are enriched within a subset of CCPs with unique lifetimes and protein composition. Perturbation of TOM1L1 or Fyn impairs EGF-stimulated phosphorylation of Akt2 but not Akt1. EGF stimulation also triggered the TOM1L1- and Fyn-dependent recruitment of the phosphoinositide 5-phosphatase SHIP2 to CCPs. Thus, the recruitment of TOM1L1 and Fyn to a subset of CCPs underlies a role for these structures in the support of EGFR signaling leading to Akt activation.
Collapse
Affiliation(s)
- Rebecca Cabral-Dias
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
| | - Stefanie Lucarelli
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
| | - Karolina Zak
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
| | - Sadia Rahmani
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
| | - Gurjeet Judge
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
| | - John Abousawan
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
| | - Laura F DiGiovanni
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Dafne Vural
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
| | - Karen E Anderson
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Michael G Sugiyama
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Gizem Genc
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Roberto J Botelho
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
| | - Gregory D Fairn
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Peter K Kim
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Reiter E. [β-arrestins, their mechanisms of action and multiple roles in the biology of G protein-coupled receptors]. Biol Aujourdhui 2022; 215:107-118. [PMID: 35275055 DOI: 10.1051/jbio/2021010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Indexed: 06/14/2023]
Abstract
The stimulation of G protein-coupled receptors (GPCRs) induces biological responses to a wide range of extracellular cues. The heterotrimeric G proteins, which are recruited to the active conformation of GPCRs, lead to the generation of various diffusible second messengers. Only two other families of proteins exhibit the remarkable characteristic of recognizing and binding to the active conformation of most GPCRs: GPCR kinases (GRKs) and β-arrestins. These two families of proteins were initially identified as key players in the desensitization of G protein activation by GPCRs. Over the years, β-arrestins have been implicated in an increasing number of interactions with non-receptor proteins, expanding the range of cellular functions in which they are involved. It is now well established that β-arrestins, by scaffolding and recruiting protein complexes in an agonist-dependent manner, directly regulate the trafficking and signaling of GPCRs. Remarkable advances have been made in recent years which have made it possible i) to identify biased ligands capable, by stabilizing particular conformations of a growing number of GPCRs, of activating or blocking the action of β-arrestins independently of that of G proteins, some of these ligands holding great therapeutic interest; ii) to demonstrate β-arrestins' role in the compartmentalization of GPCR signaling within the cell, and iii) to understand the molecular details of their interaction with GPCRs and of their activation through structural and biophysical approaches.
Collapse
Affiliation(s)
- Eric Reiter
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France - Inria, Centre de recherche Inria Saclay-Île-de-France, 91120 Palaiseau, France
| |
Collapse
|
41
|
Kolb P, Kenakin T, Alexander SPH, Bermudez M, Bohn LM, Breinholt CS, Bouvier M, Hill SJ, Kostenis E, Martemyanov K, Neubig RR, Onaran HO, Rajagopal S, Roth BL, Selent J, Shukla AK, Sommer ME, Gloriam DE. Community Guidelines for GPCR Ligand Bias: IUPHAR Review XX. Br J Pharmacol 2022; 179:3651-3674. [PMID: 35106752 PMCID: PMC7612872 DOI: 10.1111/bph.15811] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/29/2022] Open
Abstract
G protein-coupled receptors modulate a plethora of physiological processes and mediate the effects of one-third of FDA-approved drugs. Depending on which ligand activates a receptor, it can engage different intracellular transducers. This 'biased signaling' paradigm requires that we now characterize physiological signaling not just by receptors but by ligand-receptor pairs. Ligands eliciting biased signaling may constitute better drugs with higher efficacy and fewer adverse effects. However, ligand bias is very complex, making reproducibility and description challenging. Here, we provide guidelines and terminology for any scientists to design and report ligand bias experiments. The guidelines will aid consistency and clarity, as the basic receptor research and drug discovery communities continue to advance our understanding and exploitation of ligand bias. Scientific insight, biosensors, and analytical methods are still evolving and should benefit from and contribute to the implementation of the guidelines, together improving translation from in vitro to disease-relevant in vivo models.
Collapse
Affiliation(s)
- Peter Kolb
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, North, Carolina, USA
| | | | - Marcel Bermudez
- Department of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Laura M Bohn
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Christian S Breinholt
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Québec, Canada
| | - Stephen J Hill
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Evi Kostenis
- Molecular, Cellular, and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Kirill Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Rick R Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - H Ongun Onaran
- Molecular Biology and Technology Development Unit, Department of Pharmacology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Sudarshan Rajagopal
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.,Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Bryan L Roth
- Department of Pharmacology, University of North Carolina School of Medicine, North, Carolina, USA
| | - Jana Selent
- Research Programme on Biomedical Informatics, Hospital Del Mar Medical Research Institute, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| | - Martha E Sommer
- Institute of Medical Physics and Biophysics, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Current affiliation: ISAR Bioscience Institute, Munich-Planegg, Germany
| | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
Biphasic activation of β-arrestin 1 upon interaction with a GPCR revealed by methyl-TROSY NMR. Nat Commun 2021; 12:7158. [PMID: 34887409 PMCID: PMC8660791 DOI: 10.1038/s41467-021-27482-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/24/2021] [Indexed: 01/14/2023] Open
Abstract
β-arrestins (βarrs) play multifaceted roles in the function of G protein-coupled receptors (GPCRs). βarrs typically interact with phosphorylated C-terminal tail (C tail) and transmembrane core (TM core) of GPCRs. However, the effects of the C tail- and TM core-mediated interactions on the conformational activation of βarrs have remained elusive. Here, we show the conformational changes for βarr activation upon the C tail- and TM core-mediated interactions with a prototypical GPCR by nuclear magnetic resonance (NMR) spectroscopy. Our NMR analyses demonstrated that while the C tail-mediated interaction alone induces partial activation, in which βarr exists in equilibrium between basal and activated conformations, the TM core- and the C tail-mediated interactions together completely shift the equilibrium toward the activated conformation. The conformation-selective antibody, Fab30, promotes partially activated βarr into the activated-like conformation. This plasticity of βarr conformation in complex with GPCRs engaged in different binding modes may explain the multifunctionality of βarrs. β-arrestins commonly bind to two distinct elements in GPCRs: the phosphorylated carboxyl terminal tail (C tail) and the cytoplasmic face of the transmembrane region (TM core). Here, the authors use methyl-TROSY NMR measurements to characterise the interactions between β-arrestin 1 (βarr1) and a GPCR and observe that C tail-mediated interaction with a GPCR alone induces the partial activation of βarr1, whereas the TM core- and C tail-mediated interactions together stabilize the activated conformation of βarr1.
Collapse
|
43
|
Pashkova N, Gakhar L, Yu L, Schnicker NJ, Minard AY, Winistorfer S, Johnson IE, Piper RC. ANTH domains within CALM, HIP1R, and Sla2 recognize ubiquitin internalization signals. eLife 2021; 10:72583. [PMID: 34821552 PMCID: PMC8648300 DOI: 10.7554/elife.72583] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Attachment of ubiquitin (Ub) to cell surface proteins serves as a signal for internalization via clathrin-mediated endocytosis (CME). How ubiquitinated membrane proteins engage the internalization apparatus remains unclear. The internalization apparatus contains proteins such as Epsin and Eps15, which bind Ub, potentially acting as adaptors for Ub-based internalization signals. Here, we show that additional components of the endocytic machinery including CALM, HIP1R, and Sla2 bind Ub via their N-terminal ANTH domain, a domain belonging to the superfamily of ENTH and VHS domains. Structural studies revealed that Ub binds with µM affinity to a unique C-terminal region within the ANTH domain not found in ENTH domains. Functional studies showed that combined loss of Ub-binding by ANTH-domain proteins and other Ub-binding domains within the yeast internalization apparatus caused defects in the Ub-dependent internalization of the GPCR Ste2 that was engineered to rely exclusively on Ub as an internalization signal. In contrast, these mutations had no effect on the internalization of Ste2 engineered to use an alternate Ub-independent internalization signal. These studies define new components of the internalization machinery that work collectively with Epsin and Eps15 to specify recognition of Ub as an internalization signal.
Collapse
Affiliation(s)
- Natalya Pashkova
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, United States
| | - Lokesh Gakhar
- Carver College of Medicine Protein Crystallography Core, University of Iowa, Iowa City, United States.,Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, United States
| | - Liping Yu
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, United States.,Carver College of Medicine NMR Core, University of Iowa, Iowa City, United States
| | - Nicholas J Schnicker
- Carver College of Medicine Protein Crystallography Core, University of Iowa, Iowa City, United States
| | - Annabel Y Minard
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, United States
| | - Stanley Winistorfer
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, United States
| | - Ivan E Johnson
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, United States
| | - Robert C Piper
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, United States
| |
Collapse
|
44
|
Jullié D, Valbret Z, Stoeber M. Optical tools to study the subcellular organization of GPCR neuromodulation. J Neurosci Methods 2021; 366:109408. [PMID: 34763022 DOI: 10.1016/j.jneumeth.2021.109408] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/11/2021] [Accepted: 11/03/2021] [Indexed: 12/29/2022]
Abstract
Modulation of neuronal circuit activity is key to information processing in the brain. G protein-coupled receptors (GPCRs), the targets of most neuromodulatory ligands, show extremely diverse expression patterns in neurons and receptors can be localized in various sub-neuronal membrane compartments. Upon activation, GPCRs promote signaling cascades that alter the level of second messengers, drive phosphorylation changes, modulate ion channel function, and influence gene expression, all of which critically impact neuron physiology. Because of its high degree of complexity, this form of interneuronal communication has remained challenging to integrate into our conceptual understanding of brain function. Recent technological advances in fluorescence microscopy and the development of optical biosensors now allow investigating neuromodulation with unprecedented resolution on the level of individual cells. In this review, we will highlight recent imaging techniques that enable determining the precise localization of GPCRs in neurons, with specific focus on the subcellular and nanoscale level. Downstream of receptors, we describe novel conformation-specific biosensors that allow for real-time monitoring of GPCR activation and of distinct signal transduction events in neurons. Applying these new tools has the potential to provide critical insights into the function and organization of GPCRs in neuronal cells and may help decipher the molecular and cellular mechanisms that underlie neuromodulation.
Collapse
Affiliation(s)
- Damien Jullié
- Department of Psychiatry, University of California San Francisco, San Francisco, USA.
| | - Zoé Valbret
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Miriam Stoeber
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
45
|
Dendritic cell migration in inflammation and immunity. Cell Mol Immunol 2021; 18:2461-2471. [PMID: 34302064 PMCID: PMC8298985 DOI: 10.1038/s41423-021-00726-4] [Citation(s) in RCA: 262] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023] Open
Abstract
Dendritic cells (DCs) are the key link between innate immunity and adaptive immunity and play crucial roles in both the promotion of immune defense and the maintenance of immune tolerance. The trafficking of distinct DC subsets across lymphoid and nonlymphoid tissues is essential for DC-dependent activation and regulation of inflammation and immunity. DC chemotaxis and migration are triggered by interactions between chemokines and their receptors and regulated by multiple intracellular mechanisms, such as protein modification, epigenetic reprogramming, metabolic remodeling, and cytoskeletal rearrangement, in a tissue-specific manner. Dysregulation of DC migration may lead to abnormal positioning or activation of DCs, resulting in an imbalance of immune responses and even immune pathologies, including autoimmune responses, infectious diseases, allergic diseases and tumors. New strategies targeting the migration of distinct DC subsets are being explored for the treatment of inflammatory and infectious diseases and the development of novel DC-based vaccines. In this review, we will discuss the migratory routes and immunological consequences of distinct DC subsets, the molecular basis and regulatory mechanisms of migratory signaling in DCs, and the association of DC migration with the pathogenesis of autoimmune and infectious diseases.
Collapse
|
46
|
Wang W, Bian J, Sun Y, Li Z. The new fate of internalized membrane receptors: Internalized activation. Pharmacol Ther 2021; 233:108018. [PMID: 34626676 DOI: 10.1016/j.pharmthera.2021.108018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
Classically, the fate of internalized membrane receptors includes receptor degradation and receptor recycling. However, recent findings have begun to challenge these views. Much research demonstrated that many internalized membrane receptors can trigger distinct signal activation rather than being desensitized inside the cell. Here, we introduce the concept of "internalized activation" which not only represents a new mode of receptor activation, but also endows the new fate for receptor internalization (from death to life). The new activation mode and fate of membrane receptor are ubiquitous and have unique theoretical significance. We systematically put forward the features, process, and regulation of "internalized activation" and its significance in signal transduction and diseases. "Internalized activation" will provide a completely new understanding for the theory of receptor activation, internalization and novel drug targets for precision medicine.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Jingwei Bian
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Yang Sun
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Zijian Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
47
|
Constitutive signal bias mediated by the human GHRHR splice variant 1. Proc Natl Acad Sci U S A 2021; 118:2106606118. [PMID: 34599099 PMCID: PMC8501799 DOI: 10.1073/pnas.2106606118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2021] [Indexed: 11/18/2022] Open
Abstract
The mechanism of functional changes induced by alternative splicing of GHRHR is largely unknown. Here, we demonstrate that GHRH-elicited signal bias toward β-arrestin recruitment is constitutively mediated by SV1. The cryogenic electron microscopy structures of SV1 and molecular dynamics simulations reveal the different functionalities between GHRHR and SV1 at the near-atomic level (i.e., the N termini of GHRHR and SV1 differentiate the downstream signaling pathways, Gs versus β-arrestins). Our findings provide valuable insights into the functional diversity of class B1 GPCRs that may aid in the design of better therapeutic agents against certain cancers. Alternative splicing of G protein–coupled receptors has been observed, but their functions are largely unknown. Here, we report that a splice variant (SV1) of the human growth hormone–releasing hormone receptor (GHRHR) is capable of transducing biased signal. Differing only at the receptor N terminus, GHRHR predominantly activates Gs while SV1 selectively couples to β-arrestins. Based on the cryogenic electron microscopy structures of SV1 in the apo state or GHRH-bound state in complex with the Gs protein, molecular dynamics simulations reveal that the N termini of GHRHR and SV1 differentiate the downstream signaling pathways, Gs versus β-arrestins. As suggested by mutagenesis and functional studies, it appears that GHRH-elicited signal bias toward β-arrestin recruitment is constitutively mediated by SV1. The level of SV1 expression in prostate cancer cells is also positively correlated with ERK1/2 phosphorylation but negatively correlated with cAMP response. Our findings imply that constitutive signal bias may be a mechanism that ensures cancer cell proliferation.
Collapse
|
48
|
Sigismund S, Lanzetti L, Scita G, Di Fiore PP. Endocytosis in the context-dependent regulation of individual and collective cell properties. Nat Rev Mol Cell Biol 2021; 22:625-643. [PMID: 34075221 DOI: 10.1038/s41580-021-00375-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 02/07/2023]
Abstract
Endocytosis allows cells to transport particles and molecules across the plasma membrane. In addition, it is involved in the termination of signalling through receptor downmodulation and degradation. This traditional outlook has been substantially modified in recent years by discoveries that endocytosis and subsequent trafficking routes have a profound impact on the positive regulation and propagation of signals, being key for the spatiotemporal regulation of signal transmission in cells. Accordingly, endocytosis and membrane trafficking regulate virtually every aspect of cell physiology and are frequently subverted in pathological conditions. Two key aspects of endocytic control over signalling are coming into focus: context-dependency and long-range effects. First, endocytic-regulated outputs are not stereotyped but heavily dependent on the cell-specific regulation of endocytic networks. Second, endocytic regulation has an impact not only on individual cells but also on the behaviour of cellular collectives. Herein, we will discuss recent advancements in these areas, highlighting how endocytic trafficking impacts complex cell properties, including cell polarity and collective cell migration, and the relevance of these mechanisms to disease, in particular cancer.
Collapse
Affiliation(s)
- Sara Sigismund
- IEO, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Letizia Lanzetti
- Department of Oncology, University of Torino Medical School, Torino, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Torino, Italy
| | - Giorgio Scita
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy.,IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Pier Paolo Di Fiore
- IEO, European Institute of Oncology IRCCS, Milan, Italy. .,Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
49
|
Migration cues interpretation by clathrin-coated structures. Curr Opin Cell Biol 2021; 72:100-105. [PMID: 34391036 DOI: 10.1016/j.ceb.2021.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 11/24/2022]
Abstract
Cell migration is oriented by cues from the environment. Such cues are read and interpreted by the cell and translated into a reorganization of the migration machinery to steer migration. Receptors at the cell surface are central to detect these cues. These receptors can be internalized and this plays an important role in the decision-making process leading to choosing a migration direction. Independently of endocytosis, recent findings suggest that regulation of these receptors and translation of the information they carry into a phenotype is facilitated by their clustering at discrete locations of the plasma membrane. Clathrin-coated structures are archetypal clustering assemblies and thus provide the cell with a finely tunable mechanism for controlling receptor availability. In addition, clathrin-coated structures can be regulated by many factors playing a role in cell migration and thus take part in feedback loop mechanisms that are instrumental in defining a migration direction.
Collapse
|
50
|
Jean-Alphonse FG, Sposini S. Confocal and TIRF microscopy based approaches to visualize arrestin trafficking in living cells. Methods Cell Biol 2021; 166:179-203. [PMID: 34752332 DOI: 10.1016/bs.mcb.2021.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Arrestins are key proteins that serve as versatile scaffolds to control and mediate G protein coupled receptors (GPCR) activity. Arrestin control of GPCR functions involves their recruitment from the cytosol to plasma membrane-localized GPCRs and to endosomal compartments, where they mediate internalization, sorting and signaling of GPCRs. Several methods can be used to monitor trafficking of arrestins; however, live fluorescence imaging remains the method of choice to both assess arrestin recruitment to ligand-activated receptors and to monitor its dynamic subcellular localization. Here, we present two approaches based on Total Internal Fluorescence (TIRF) microscopy and confocal microscopy to visualize arrestin trafficking in live cells in real time and to assess their co-localization with the GPCR of interest and their localization at specific subcellular locations.
Collapse
Affiliation(s)
- Frédéric Gaëtan Jean-Alphonse
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France; Université Paris-Saclay, Inria, Inria Saclay-Île-de-France, Palaiseau, France
| | - Silvia Sposini
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom; University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, Bordeaux, France.
| |
Collapse
|