1
|
Matthees ESF, Filor JC, Jaiswal N, Reichel M, Youssef N, D'Uonnolo G, Szpakowska M, Drube J, König GM, Kostenis E, Chevigné A, Godbole A, Hoffmann C. GRK specificity and Gβγ dependency determines the potential of a GPCR for arrestin-biased agonism. Commun Biol 2024; 7:802. [PMID: 38956302 PMCID: PMC11220067 DOI: 10.1038/s42003-024-06490-1] [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: 12/15/2023] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are mainly regulated by GPCR kinase (GRK) phosphorylation and subsequent β-arrestin recruitment. The ubiquitously expressed GRKs are classified into cytosolic GRK2/3 and membrane-tethered GRK5/6 subfamilies. GRK2/3 interact with activated G protein βγ-subunits to translocate to the membrane. Yet, this need was not linked as a factor for bias, influencing the effectiveness of β-arrestin-biased agonist creation. Using multiple approaches such as GRK2/3 mutants unable to interact with Gβγ, membrane-tethered GRKs and G protein inhibitors in GRK2/3/5/6 knockout cells, we show that G protein activation will precede GRK2/3-mediated β-arrestin2 recruitment to activated receptors. This was independent of the source of free Gβγ and observable for Gs-, Gi- and Gq-coupled GPCRs. Thus, β-arrestin interaction for GRK2/3-regulated receptors is inseparably connected with G protein activation. We outline a theoretical framework of how GRK dependence on free Gβγ can determine a GPCR's potential for biased agonism. Due to this inherent cellular mechanism for GRK2/3 recruitment and receptor phosphorylation, we anticipate generation of β-arrestin-biased ligands to be mechanistically challenging for the subgroup of GPCRs exclusively regulated by GRK2/3, but achievable for GRK5/6-regulated receptors, that do not demand liberated Gβγ. Accordingly, GRK specificity of any GPCR is foundational for developing arrestin-biased ligands.
Collapse
Affiliation(s)
- Edda S F Matthees
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine; Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - Jenny C Filor
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine; Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - Natasha Jaiswal
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine; Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Mona Reichel
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine; Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - Noureldine Youssef
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine; Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - Giulia D'Uonnolo
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Martyna Szpakowska
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg
| | - Julia Drube
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine; Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - Gabriele M König
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, D-53115, Bonn, Germany
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, D-53115, Bonn, Germany
| | - Andy Chevigné
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg
| | - Amod Godbole
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine; Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - Carsten Hoffmann
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine; Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany.
| |
Collapse
|
2
|
McNeill SM, Zhao P. The roles of RGS proteins in cardiometabolic disease. Br J Pharmacol 2024; 181:2319-2337. [PMID: 36964984 DOI: 10.1111/bph.16076] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/12/2023] [Accepted: 03/20/2023] [Indexed: 03/27/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are the most prominent receptors on the surface of the cell and play a central role in the regulation of cardiac and metabolic functions. GPCRs transmit extracellular stimuli to the interior of the cells by activating one or more heterotrimeric G proteins. The duration and intensity of G protein-mediated signalling are tightly controlled by a large array of intracellular mediators, including the regulator of G protein signalling (RGS) proteins. RGS proteins selectively promote the GTPase activity of a subset of Gα subunits, thus serving as negative regulators in a pathway-dependent manner. In the current review, we summarise the involvement of RGS proteins in cardiometabolic function with a focus on their tissue distribution, mechanisms of action and dysregulation under various disease conditions. We also discuss the potential therapeutic applications for targeting RGS proteins in treating cardiometabolic conditions and current progress in developing RGS modulators. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Samantha M McNeill
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Peishen Zhao
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins (CCeMMP), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
3
|
Tóth AD, Szalai B, Kovács OT, Garger D, Prokop S, Soltész-Katona E, Balla A, Inoue A, Várnai P, Turu G, Hunyady L. G protein-coupled receptor endocytosis generates spatiotemporal bias in β-arrestin signaling. Sci Signal 2024; 17:eadi0934. [PMID: 38917219 DOI: 10.1126/scisignal.adi0934] [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: 04/03/2023] [Accepted: 06/05/2024] [Indexed: 06/27/2024]
Abstract
The stabilization of different active conformations of G protein-coupled receptors is thought to underlie the varying efficacies of biased and balanced agonists. Here, profiling the activation of signal transducers by angiotensin II type 1 receptor (AT1R) agonists revealed that the extent and kinetics of β-arrestin binding exhibited substantial ligand-dependent differences, which were lost when receptor internalization was inhibited. When AT1R endocytosis was prevented, even weak partial agonists of the β-arrestin pathway acted as full or near-full agonists, suggesting that receptor conformation did not exclusively determine β-arrestin recruitment. The ligand-dependent variance in β-arrestin translocation was much larger at endosomes than at the plasma membrane, showing that ligand efficacy in the β-arrestin pathway was spatiotemporally determined. Experimental investigations and mathematical modeling demonstrated how multiple factors concurrently shaped the effects of agonists on endosomal receptor-β-arrestin binding and thus determined the extent of functional selectivity. Ligand dissociation rate and G protein activity had particularly strong, internalization-dependent effects on the receptor-β-arrestin interaction. We also showed that endocytosis regulated the agonist efficacies of two other receptors with sustained β-arrestin binding: the V2 vasopressin receptor and a mutant β2-adrenergic receptor. In the absence of endocytosis, the agonist-dependent variance in β-arrestin2 binding was markedly diminished. Our results suggest that endocytosis determines the spatiotemporal bias in GPCR signaling and can aid in the development of more efficacious, functionally selective compounds.
Collapse
MESH Headings
- Endocytosis/physiology
- Humans
- Signal Transduction
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/genetics
- beta-Arrestins/metabolism
- beta-Arrestins/genetics
- HEK293 Cells
- Receptors, Vasopressin/metabolism
- Receptors, Vasopressin/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, Adrenergic, beta-2/genetics
- Endosomes/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Animals
- Ligands
- Protein Binding
- Protein Transport
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, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, Tűzoltó utca 37-47, H-1094 Budapest, Hungary
- Department of Internal Medicine and Haematology, Semmelweis University, Szentkirályi utca 46, H-1088 Budapest, Hungary
| | - Bence Szalai
- Institute of Molecular Life Sciences, Centre of Excellence of the Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, Tűzoltó utca 37-47, H-1094 Budapest, Hungary
| | - Orsolya T Kovács
- Department of Physiology, Faculty of Medicine, Semmelweis University, Tűzoltó utca 37-47, H-1094 Budapest, Hungary
| | - Dániel Garger
- Department of Physiology, Faculty of Medicine, Semmelweis University, Tűzoltó utca 37-47, H-1094 Budapest, Hungary
- Computational Health Center, Helmholtz Munich, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany
| | - Susanne Prokop
- Department of Physiology, Faculty of Medicine, Semmelweis University, Tűzoltó utca 37-47, H-1094 Budapest, Hungary
| | - Eszter Soltész-Katona
- Institute of Molecular Life Sciences, Centre of Excellence of the Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - András Balla
- Department of Physiology, Faculty of Medicine, Semmelweis University, Tűzoltó utca 37-47, H-1094 Budapest, Hungary
- HUN-REN-SE Laboratory of Molecular Physiology, Hungarian Research Network, Tűzoltó utca 37-47, H-1094 Budapest, Hungary
| | - Asuka Inoue
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578 Japan
| | - Péter Várnai
- Department of Physiology, Faculty of Medicine, Semmelweis University, Tűzoltó utca 37-47, H-1094 Budapest, Hungary
- HUN-REN-SE Laboratory of Molecular Physiology, Hungarian Research Network, Tűzoltó utca 37-47, H-1094 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, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, Tűzoltó utca 37-47, H-1094 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, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, Tűzoltó utca 37-47, H-1094 Budapest, Hungary
| |
Collapse
|
4
|
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
|
5
|
Hammill AM, Boscolo E. Capillary malformations. J Clin Invest 2024; 134:e172842. [PMID: 38618955 PMCID: PMC11014659 DOI: 10.1172/jci172842] [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: 04/16/2024] Open
Abstract
Capillary malformation (CM), or port wine birthmark, is a cutaneous congenital vascular anomaly that occurs in 0.1%-2% of newborns. Patients with a CM localized on the forehead have an increased risk of developing a neurocutaneous disorder called encephalotrigeminal angiomatosis or Sturge-Weber syndrome (SWS), with complications including seizure, developmental delay, glaucoma, and vision loss. In 2013, a groundbreaking study revealed causative activating somatic mutations in the gene (GNAQ) encoding guanine nucleotide-binding protein Q subunit α (Gαq) in CM and SWS patient tissues. In this Review, we discuss the disease phenotype, the causative GNAQ mutations, and their cellular origin. We also present the endothelial Gαq-related signaling pathways, the current animal models to study CM and its complications, and future options for therapeutic treatment. Further work remains to fully elucidate the cellular and molecular mechanisms underlying the formation and maintenance of the abnormal vessels.
Collapse
Affiliation(s)
- Adrienne M. Hammill
- Division of Hematology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Elisa Boscolo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
6
|
Wu C, Hu L, Liu B, Zeng X, Ma H, Cao Y, Li H, Zhang X. TRAF6-mediated ubiquitination of AKT in the nucleus is a critical event underlying the desensitization of G protein-coupled receptors. Cell Commun Signal 2024; 22:213. [PMID: 38566235 PMCID: PMC10986131 DOI: 10.1186/s12964-024-01592-z] [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: 01/21/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Desensitization of G protein-coupled receptors (GPCRs) refers to the attenuation of receptor responsiveness by prolonged or intermittent exposure to agonists. The binding of β-arrestin to the cytoplasmic cavity of the phosphorylated receptor, which competes with the G protein, has been widely accepted as an extensive model for explaining GPCRs desensitization. However, studies on various GPCRs, including dopamine D2-like receptors (D2R, D3R, D4R), have suggested the existence of other desensitization mechanisms. The present study employed D2R/D3R variants with different desensitization properties and utilized loss-of-function approaches to uncover the mechanisms underlying GPCRs homologous desensitization, focusing on the signaling cascade that regulates the ubiquitination of AKT. RESULTS AKT undergoes K8/14 ubiquitination by TRAF6, which occurs in the nucleus and promotes its membrane recruitment, phosphorylation and activation under receptor desensitization conditions. The nuclear entry of TRAF6 relies on the presence of the importin complex. Src regulates the nuclear entry of TRAF6 by mediating the interaction between TRAF6 and importin β1. Ubiquitinated AKT translocates to the plasma membrane where it associates with Mdm2 to phosphorylate it at the S166 and S186 residues. Thereafter, phosphorylated Mdm2 is recruited to the nucleus, resulting in the deubiquitination of β-Arr2. The deubiquitinated β-Arr2 then forms a complex with Gβγ, which serves as a biomarker for GPCRs desensitization. Like in D3R, ubiquitination of AKT is also involved in the desensitization of β2 adrenoceptors. CONCLUSION Our study proposed that the property of a receptor that causes a change in the subcellular localization of TRAF6 from the cytoplasm to the nucleus to mediate AKT ubiquitination could initiate the desensitization of GPCRs.
Collapse
Affiliation(s)
- Chengyan Wu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
| | - Li Hu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
| | - Bing Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
| | - Xingyue Zeng
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
| | - Haixiang Ma
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
| | - Yongkai Cao
- Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Huijun Li
- Department of Pharmaceuticals, People's Hospital of Zunyi City Bo Zhou District, Zunyi, 563000, China
| | - Xiaohan Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
7
|
Varney MJ, Benovic JL. The Role of G Protein-Coupled Receptors and Receptor Kinases in Pancreatic β-Cell Function and Diabetes. Pharmacol Rev 2024; 76:267-299. [PMID: 38351071 PMCID: PMC10877731 DOI: 10.1124/pharmrev.123.001015] [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] [Received: 08/03/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 02/16/2024] Open
Abstract
Type 2 diabetes (T2D) mellitus has emerged as a major global health concern that has accelerated in recent years due to poor diet and lifestyle. Afflicted individuals have high blood glucose levels that stem from the inability of the pancreas to make enough insulin to meet demand. Although medication can help to maintain normal blood glucose levels in individuals with chronic disease, many of these medicines are outdated, have severe side effects, and often become less efficacious over time, necessitating the need for insulin therapy. G protein-coupled receptors (GPCRs) regulate many physiologic processes, including blood glucose levels. In pancreatic β cells, GPCRs regulate β-cell growth, apoptosis, and insulin secretion, which are all critical in maintaining sufficient β-cell mass and insulin output to ensure euglycemia. In recent years, new insights into the signaling of incretin receptors and other GPCRs have underscored the potential of these receptors as desirable targets in the treatment of diabetes. The signaling of these receptors is modulated by GPCR kinases (GRKs) that phosphorylate agonist-activated GPCRs, marking the receptor for arrestin binding and internalization. Interestingly, genome-wide association studies using diabetic patient cohorts link the GRKs and arrestins with T2D. Moreover, recent reports show that GRKs and arrestins expressed in the β cell serve a critical role in the regulation of β-cell function, including β-cell growth and insulin secretion in both GPCR-dependent and -independent pathways. In this review, we describe recent insights into GPCR signaling and the importance of GRK function in modulating β-cell physiology. SIGNIFICANCE STATEMENT: Pancreatic β cells contain a diverse array of G protein-coupled receptors (GPCRs) that have been shown to improve β-cell function and survival, yet only a handful have been successfully targeted in the treatment of diabetes. This review discusses recent advances in our understanding of β-cell GPCR pharmacology and regulation by GPCR kinases while also highlighting the necessity of investigating islet-enriched GPCRs that have largely been unexplored to unveil novel treatment strategies.
Collapse
Affiliation(s)
- Matthew J Varney
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
8
|
Jiang W, Wang W, Kong Y, Zheng S. Structural basis for the ubiquitination of G protein βγ subunits by KCTD5/Cullin3 E3 ligase. SCIENCE ADVANCES 2023; 9:eadg8369. [PMID: 37450587 PMCID: PMC10348674 DOI: 10.1126/sciadv.adg8369] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
G protein-coupled receptor (GPCR) signaling is precisely controlled to avoid overstimulation that results in detrimental consequences. Gβγ signaling is negatively regulated by a Cullin3 (Cul3)-dependent E3 ligase, KCTD5, which triggers ubiquitination and degradation of free Gβγ. Here, we report the cryo-electron microscopy structures of the KCTD5-Gβγ fusion complex and the KCTD7-Cul3 complex. KCTD5 in pentameric form engages symmetrically with five copies of Gβγ through its C-terminal domain. The unique pentameric assembly of the KCTD5/Cul3 E3 ligase places the ubiquitin-conjugating enzyme (E2) and the modification sites of Gβγ in close proximity and allows simultaneous transfer of ubiquitin from E2 to five Gβγ subunits. Moreover, we show that ubiquitination of Gβγ by KCTD5 is important for fine-tuning cyclic adenosine 3´,5´-monophosphate signaling of GPCRs. Our studies provide unprecedented insights into mechanisms of substrate recognition by unusual pentameric E3 ligases and highlight the KCTD family as emerging regulators of GPCR signaling.
Collapse
Affiliation(s)
- Wentong Jiang
- Graduate School of Peking Union Medical College, Beijing 100730, China
- National Institute of Biological Sciences, Beijing 102206, China
| | - Wei Wang
- National Institute of Biological Sciences, Beijing 102206, China
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Yinfei Kong
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Sanduo Zheng
- Graduate School of Peking Union Medical College, Beijing 100730, China
- National Institute of Biological Sciences, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
9
|
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: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
10
|
Xiang G, Acosta-Ruiz A, Radoux-Mergault A, Kristt M, Kim J, Moon JD, Broichhagen J, Inoue A, Lee FS, Stoeber M, Dittman JS, Levitz J. Control of Gα q signaling dynamics and GPCR cross-talk by GRKs. SCIENCE ADVANCES 2022; 8:eabq3363. [PMID: 36427324 PMCID: PMC9699688 DOI: 10.1126/sciadv.abq3363] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Numerous processes contribute to the regulation of G protein-coupled receptors (GPCRs), but relatively little is known about rapid mechanisms that control signaling on the seconds time scale or regulate cross-talk between receptors. Here, we reveal that the ability of some GPCR kinases (GRKs) to bind Gαq both drives acute signaling desensitization and regulates functional interactions between GPCRs. GRK2/3-mediated acute desensitization occurs within seconds, is rapidly reversible, and can occur upon local, subcellular activation. This rapid desensitization is kinase independent, insensitive to pharmacological inhibition, and generalizable across receptor families and effectors. We also find that the ability of GRK2 to bind G proteins also enables it to regulate the extent and timing of Gαq-dependent signaling cross-talk between GPCRs. Last, we find that G protein/GRK2 interactions enable a novel form of GPCR trafficking cross-talk. Together, this work reveals potent forms of Gαq-dependent GPCR regulation with wide-ranging pharmacological and physiological implications.
Collapse
Affiliation(s)
- Guoqing Xiang
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Melanie Kristt
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Jihye Kim
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Jared D. Moon
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | | | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Francis S. Lee
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Miriam Stoeber
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Jeremy S. Dittman
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
- Corresponding author.
| |
Collapse
|
11
|
Randolph CE, Dwyer MB, Aumiller JL, Dixon AJ, Inoue A, Osei-Owusu P, Wedegaertner PB. Enhanced membrane binding of oncogenic G protein αqQ209L confers resistance to inhibitor YM-254890. J Biol Chem 2022; 298:102538. [PMID: 36174676 PMCID: PMC9626947 DOI: 10.1016/j.jbc.2022.102538] [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: 03/24/2022] [Revised: 09/15/2022] [Accepted: 09/18/2022] [Indexed: 11/29/2022] Open
Abstract
Heterotrimeric G proteins couple activated G protein–coupled receptors (GPCRs) to intracellular signaling pathways. They can also function independently of GPCR activation upon acquiring mutations that prevent GTPase activity and result in constitutive signaling, as occurs with the αqQ209L mutation in uveal melanoma. YM-254890 (YM) can inhibit signaling by both GPCR-activated WT αq and GPCR-independent αqQ209L. Although YM inhibits WT αq by binding to αq-GDP and preventing GDP/GTP exchange, the mechanism of YM inhibition of cellular αqQ209L remains to be fully understood. Here, we show that YM promotes a subcellular redistribution of αqQ209L from the plasma membrane (PM) to the cytoplasm. To test if this loss of PM localization could contribute to the mechanism of inhibition of αqQ209L by YM, we developed and examined N-terminal mutants of αqQ209L, termed PM-restricted αqQ209L, in which the addition of membrane-binding motifs enhanced PM localization and prevented YM-promoted redistribution. Treatment of cells with YM failed to inhibit signaling by these PM-restricted αqQ209L. Additionally, pull-down experiments demonstrated that YM promotes similar conformational changes in both αqQ209L and PM-restricted αqQ209L, resulting in increased binding to βγ and decreased binding to regulator RGS2, and effectors p63RhoGEF-DH/PH and phospholipase C-β. GPCR-dependent signaling by PM-restricted WT αq is strongly inhibited by YM, demonstrating that resistance to YM inhibition by membrane-binding mutants is specific to constitutively active αqQ209L. Together, these results indicate that changes in membrane binding impact the ability of YM to inhibit αqQ209L and suggest that YM contributes to inhibition of αqQ209L by promoting its relocalization.
Collapse
Affiliation(s)
- Clinita E Randolph
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Morgan B Dwyer
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Jenna L Aumiller
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Alethia J Dixon
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Patrick Osei-Owusu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA.
| |
Collapse
|
12
|
Jiang H, Galtes D, Wang J, Rockman HA. G protein-coupled receptor signaling: transducers and effectors. Am J Physiol Cell Physiol 2022; 323:C731-C748. [PMID: 35816644 PMCID: PMC9448338 DOI: 10.1152/ajpcell.00210.2022] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/27/2022] [Accepted: 07/10/2022] [Indexed: 01/14/2023]
Abstract
G protein-coupled receptors (GPCRs) are of considerable interest due to their importance in a wide range of physiological functions and in a large number of Food and Drug Administration (FDA)-approved drugs as therapeutic entities. With continued study of their function and mechanism of action, there is a greater understanding of how effector molecules interact with a receptor to initiate downstream effector signaling. This review aims to explore the signaling pathways, dynamic structures, and physiological relevance in the cardiovascular system of the three most important GPCR signaling effectors: heterotrimeric G proteins, GPCR kinases (GRKs), and β-arrestins. We will first summarize their prominent roles in GPCR pharmacology before transitioning into less well-explored areas. As new technologies are developed and applied to studying GPCR structure and their downstream effectors, there is increasing appreciation for the elegance of the regulatory mechanisms that mediate intracellular signaling and function.
Collapse
Affiliation(s)
- Haoran Jiang
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Daniella Galtes
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Jialu Wang
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
13
|
Zhai R, Snyder J, Montgomery S, Sato PY. Double life: How GRK2 and β-arrestin signaling participate in diseases. Cell Signal 2022; 94:110333. [PMID: 35430346 PMCID: PMC9929935 DOI: 10.1016/j.cellsig.2022.110333] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 11/03/2022]
Abstract
G-protein coupled receptor (GPCR) kinases (GRKs) and β-arrestins play key roles in GPCR and non-GPCR cellular responses. In fact, GRKs and arrestins are involved in a plethora of pathways vital for physiological maintenance of inter- and intracellular communication. Here we review decades of research literature spanning from the discovery, identification of key structural elements, and findings supporting the diverse roles of these proteins in GPCR-mediated pathways. We then describe how GRK2 and β-arrestins partake in non-GPCR signaling and briefly summarize their involvement in various pathologies. We conclude by presenting gaps in knowledge and our prospective on the promising pharmacological potential in targeting these proteins and/or downstream signaling. Future research is warranted and paramount for untangling these novel and promising roles for GRK2 and arrestins in metabolism and disease progression.
Collapse
Affiliation(s)
| | | | | | - Priscila Y. Sato
- Corresponding author at: Drexel University College of Medicine, Department of Pharmacology and Physiology, 245 N 15th Street, NCB 8152, Philadelphia, PA 19102, USA. (P.Y. Sato)
| |
Collapse
|
14
|
Echeverría E, Ripoll S, Fabián L, Shayo C, Monczor F, Fernández NC. Novel inhibitors of phosphorylation independent activity of GRK2 modulate cAMP signaling. Pharmacol Res Perspect 2022; 10:e00913. [PMID: 35184416 PMCID: PMC8858223 DOI: 10.1002/prp2.913] [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: 10/20/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/10/2022] Open
Abstract
G protein-coupled receptors kinase 2 (GRK2) plays a major role in receptor regulation and, as a consequence, in cell biology and physiology. GRK2-mediated receptor desensitization is performed by its kinase domain, which exerts receptor phosphorylation promoting G protein uncoupling and the cessation of signaling, and by its RGS homology (RH) domain, able to interrupt G protein signaling. Since GRK2 activity is exacerbated in several pathologies, many efforts to develop inhibitors have been conducted. Most of them were directed toward GRK2 kinase activity and showed encouraging results on in vitro systems and animal models. Nevertheless, limitations including unspecific effects or pharmacokinetics issues prevented them from advancing to clinical trials. Surprisingly, even though the RH domain demonstrated the ability to desensitize GPCRs, this domain has been less explored. Herein, we show in vitro activity of a series of compounds that, by inhibiting GRK2 RH domain, increase receptor cAMP response, avoid GRK2 translocation to the plasma membrane, inhibit coimmunoprecipitation of GRK2 with Gαs subunit of heterotrimeric G protein, and prevent receptor desensitization. Also, we preliminarily evaluated candidates' ADMET properties and observed suitable lipophilicity and cytotoxicity. These novel inhibitors of phosphorylation-independent actions of GRK2 might be useful in elucidating other RH domain roles and lay the foundation for the development of innovative pharmacologic therapy for diseases where GRK2 activity is exacerbated.
Collapse
Affiliation(s)
- Emiliana Echeverría
- Facultad de Farmacia y BioquímicaUniversidad de Buenos AiresBuenos AiresArgentina
- Instituto de Investigaciones Farmacológicas (ININFA‐UBA‐CONICET)Facultad de Farmacia y BioquímicaUniversidad de Buenos AiresBuenos AiresArgentina
| | - Sonia Ripoll
- Facultad de Farmacia y BioquímicaUniversidad de Buenos AiresBuenos AiresArgentina
- Instituto de Investigaciones Farmacológicas (ININFA‐UBA‐CONICET)Facultad de Farmacia y BioquímicaUniversidad de Buenos AiresBuenos AiresArgentina
| | - Lucas Fabián
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA‐UBA‐CONICET)Facultad de Farmacia y BioquímicaUniversidad de Buenos AiresBuenos AiresArgentina
| | - Carina Shayo
- Laboratorio de Patología y Farmacología MolecularInstituto de Biología y Medicina Experimental (IByME)CONICETBuenos AiresArgentina
| | - Federico Monczor
- Facultad de Farmacia y BioquímicaUniversidad de Buenos AiresBuenos AiresArgentina
- Instituto de Investigaciones Farmacológicas (ININFA‐UBA‐CONICET)Facultad de Farmacia y BioquímicaUniversidad de Buenos AiresBuenos AiresArgentina
| | - Natalia C. Fernández
- Facultad de Farmacia y BioquímicaUniversidad de Buenos AiresBuenos AiresArgentina
- Instituto de Investigaciones Farmacológicas (ININFA‐UBA‐CONICET)Facultad de Farmacia y BioquímicaUniversidad de Buenos AiresBuenos AiresArgentina
| |
Collapse
|
15
|
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: 92] [Impact Index Per Article: 46.0] [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
|
16
|
Nagai J, Bellafard A, Qu Z, Yu X, Ollivier M, Gangwani MR, Diaz-Castro B, Coppola G, Schumacher SM, Golshani P, Gradinaru V, Khakh BS. Specific and behaviorally consequential astrocyte G q GPCR signaling attenuation in vivo with iβARK. Neuron 2021; 109:2256-2274.e9. [PMID: 34139149 PMCID: PMC8418870 DOI: 10.1016/j.neuron.2021.05.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/14/2021] [Accepted: 05/18/2021] [Indexed: 12/25/2022]
Abstract
Astrocytes respond to neurotransmitters and neuromodulators using G-protein-coupled receptors (GPCRs) to mediate physiological responses. Despite their importance, there has been no method to genetically, specifically, and effectively attenuate astrocyte Gq GPCR pathways to explore consequences of this prevalent signaling mechanism in vivo. We report a 122-residue inhibitory peptide from β-adrenergic receptor kinase 1 (iβARK; and inactive D110A control) to attenuate astrocyte Gq GPCR signaling. iβARK significantly attenuated Gq GPCR Ca2+ signaling in brain slices and, in vivo, altered behavioral responses, spared other GPCR responses, and did not alter astrocyte spontaneous Ca2+ signals, morphology, electrophysiological properties, or gene expression in the striatum. Furthermore, brain-wide attenuation of astrocyte Gq GPCR signaling with iβARK using PHP.eB adeno-associated viruses (AAVs), when combined with c-Fos mapping, suggested nuclei-specific contributions to behavioral adaptation and spatial memory. iβARK extends the toolkit needed to explore functions of astrocyte Gq GPCR signaling within neural circuits in vivo.
Collapse
Affiliation(s)
- Jun Nagai
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; RIKEN Center for Brain Science, 2-1 Hirosawa Wako City, Saitama 351-0198, Japan
| | - Arash Bellafard
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Zhe Qu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Xinzhu Yu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, 514 Burrill Hall, 407 S. Goodwin Ave, Urbana, IL 61801, USA
| | - Matthias Ollivier
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Mohitkumar R Gangwani
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Blanca Diaz-Castro
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Giovanni Coppola
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Sarah M Schumacher
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Peyman Golshani
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; West LA Veterans Affairs Medical Center, Los Angeles, CA 90073, USA; Intellectual and Developmental Disabilities Research Center, Los Angeles, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, University of California, David Geffen School of Medicine, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
17
|
Han C, Li Y, Zhang Y, Wang Y, Cui D, Luo T, Zhang Y, Liu Q, Li H, Wang C, Xu D, Ma Y, Wei W. Targeted inhibition of GRK2 kinase domain by CP-25 to reverse fibroblast-like synoviocytes dysfunction and improve collagen-induced arthritis in rats. Acta Pharm Sin B 2021; 11:1835-1852. [PMID: 34386323 PMCID: PMC8343125 DOI: 10.1016/j.apsb.2021.01.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/28/2020] [Accepted: 01/06/2021] [Indexed: 12/23/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease and is mainly characterized by abnormal proliferation of fibroblast-like synoviocytes (FLS). The up-regulated cellular membrane expression of G protein coupled receptor kinase 2 (GRK2) of FLS plays a critical role in RA progression, the increase of GRK2 translocation activity promotes dysfunctional prostaglandin E4 receptor (EP4) signaling and FLS abnormal proliferation. Recently, although our group found that paeoniflorin-6ʹ-O-benzene sulfonate (CP-25), a novel compound, could reverse FLS dysfunction via GRK2, little is known as to how GRK2 translocation activity is suppressed. Our findings revealed that GRK2 expression up-regulated and EP4 expression down-regulated in synovial tissues of RA patients and collagen-induced arthritis (CIA) rats, and prostaglandin E2 (PGE2) level increased in arthritis. CP-25 could down-regulate GRK2 expression, up-regulate EP4 expression, and improve synovitis of CIA rats. CP-25 and GRK2 inhibitors (paroxetine or GSK180736A) inhibited the abnormal proliferation of FLS in RA patients and CIA rats by down-regulating GRK2 translocation to EP4 receptor. The results of microscale thermophoresis (MST), cellular thermal shift assay, and inhibition of kinase activity assay indicated that CP-25 could directly target GRK2, increase the protein stability of GRK2 in cells, and inhibit GRK2 kinase activity. The docking of CP-25 and GRK2 suggested that the kinase domain of GRK2 might be an important active pocket for CP-25. G201, K220, K230, A321, and D335 in kinase domain of GRK2 might form hydrogen bonds with CP-25. Site-directed mutagenesis and co-immunoprecipitation assay further revealed that CP-25 down-regulated the interaction of GRK2 and EP4 via controlling the key amino acid residue of Ala321 of GRK2. Our data demonstrate that FLS proliferation is regulated by GRK2 translocation to EP4. Targeted inhibition of GRK2 kinase domain by CP-25 improves FLS function and represents an innovative drug for the treatment of RA by targeting GRK2.
Collapse
Affiliation(s)
- Chenchen Han
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
- Public Health and Preventive Medicine Postdoctoral Research Station of Anhui Medical University, Hefei 230032, China
| | - Yifan Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Yuwen Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Yang Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Dongqian Cui
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Tingting Luo
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Yu Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Qian Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Hao Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Chun Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Dexiang Xu
- Public Health and Preventive Medicine Postdoctoral Research Station of Anhui Medical University, Hefei 230032, China
- Corresponding authors. Tel./fax: +86 551 65161209.
| | - Yang Ma
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
- Corresponding authors. Tel./fax: +86 551 65161209.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
- Corresponding authors. Tel./fax: +86 551 65161209.
| |
Collapse
|
18
|
BRET-based effector membrane translocation assay monitors GPCR-promoted and endocytosis-mediated G q activation at early endosomes. Proc Natl Acad Sci U S A 2021; 118:2025846118. [PMID: 33990469 DOI: 10.1073/pnas.2025846118] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are gatekeepers of cellular homeostasis and the targets of a large proportion of drugs. In addition to their signaling activity at the plasma membrane, it has been proposed that their actions may result from translocation and activation of G proteins at endomembranes-namely endosomes. This could have a significant impact on our understanding of how signals from GPCR-targeting drugs are propagated within the cell. However, little is known about the mechanisms that drive G protein movement and activation in subcellular compartments. Using bioluminescence resonance energy transfer (BRET)-based effector membrane translocation assays, we dissected the mechanisms underlying endosomal Gq trafficking and activity following activation of Gq-coupled receptors, including the angiotensin II type 1, bradykinin B2, oxytocin, thromboxane A2 alpha isoform, and muscarinic acetylcholine M3 receptors. Our data reveal that GPCR-promoted activation of Gq at the plasma membrane induces its translocation to endosomes independently of β-arrestin engagement and receptor endocytosis. In contrast, Gq activity at endosomes was found to rely on both receptor endocytosis-dependent and -independent mechanisms. In addition to shedding light on the molecular processes controlling subcellular Gq signaling, our study provides a set of tools that will be generally applicable to the study of G protein translocation and activation at endosomes and other subcellular organelles, as well as the contribution of signal propagation to drug action.
Collapse
|
19
|
Kankanamge D, Ubeysinghe S, Tennakoon M, Pantula PD, Mitra K, Giri L, Karunarathne A. Dissociation of the G protein βγ from the Gq-PLCβ complex partially attenuates PIP2 hydrolysis. J Biol Chem 2021; 296:100702. [PMID: 33901492 PMCID: PMC8138763 DOI: 10.1016/j.jbc.2021.100702] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 04/09/2021] [Accepted: 04/21/2021] [Indexed: 01/14/2023] Open
Abstract
Phospholipase C β (PLCβ), which is activated by the Gq family of heterotrimeric G proteins, hydrolyzes the inner membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2), generating diacylglycerol and inositol 1,4,5-triphosphate (IP3). Because Gq and PLCβ regulate many crucial cellular processes and have been identified as major disease drivers, activation and termination of PLCβ signaling by the Gαq subunit have been extensively studied. Gq-coupled receptor activation induces intense and transient PIP2 hydrolysis, which subsequently recovers to a low-intensity steady-state equilibrium. However, the molecular underpinnings of this equilibrium remain unclear. Here, we explored the influence of signaling crosstalk between Gq and Gi/o pathways on PIP2 metabolism in living cells using single-cell and optogenetic approaches to spatially and temporally constrain signaling. Our data suggest that the Gβγ complex is a component of the highly efficient lipase GαqGTP-PLCβ-Gβγ. We found that over time, Gβγ dissociates from this lipase complex, leaving the less-efficient GαqGTP-PLCβ lipase complex and allowing the significant partial recovery of PIP2 levels. Our findings also indicate that the subtype of the Gγ subunit in Gβγ fine-tunes the lipase activity of Gq-PLCβ, in which cells expressing Gγ with higher plasma membrane interaction show lower PIP2 recovery. Given that Gγ shows cell- and tissue-specific subtype expression, our findings suggest the existence of tissue-specific distinct Gq-PLCβ signaling paradigms. Furthermore, these results also outline a molecular process that likely safeguards cells from excessive Gq signaling.
Collapse
Affiliation(s)
- Dinesh Kankanamge
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA
| | - Sithurandi Ubeysinghe
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA
| | - Mithila Tennakoon
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA
| | - Priyanka Devi Pantula
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, India
| | - Kishalay Mitra
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, India
| | - Lopamudra Giri
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, India
| | - Ajith Karunarathne
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA.
| |
Collapse
|
20
|
Mechanistic diversity involved in the desensitization of G protein-coupled receptors. Arch Pharm Res 2021; 44:342-353. [PMID: 33761113 DOI: 10.1007/s12272-021-01320-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/14/2021] [Indexed: 01/14/2023]
Abstract
The desensitization of G protein-coupled receptors (GPCRs), which involves rapid loss of responsiveness due to repeated or chronic exposure to agonists, can occur through various mechanisms at different levels of signaling pathways. In this review, the mechanisms of GPCR desensitization are classified according to their occurrence at the receptor level and downstream to the receptor. The desensitization at the receptor level occurs in a phosphorylation-dependent manner, wherein the activated receptors are phosphorylated by GPCR kinases (GRKs), thereby increasing their affinities for arrestins. Arrestins bind to receptors through the cavity on the cytoplasmic region of heptahelical domains and interfere with the binding and activation of G-protein. Diverse mechanisms are involved in the desensitization that occurs downstream of the receptor. Some of these include the sequestration of G proteins, such as Gq and Gi/o by GRK2/3 and deubiquitinated arrestins, respectively. Mechanistically, GRK2/3 attenuates GPCR signaling by sequestering the Gα subunits of the Gq family and Gβγ via regulators of G protein signaling and pleckstrin homology domains, respectively. Moreover, studies on Gi/o-coupled D2-like receptors have reported that arrestins are deubiquitinated under desensitization condition and form a stable complex with Gβγ, thereby preventing them from coupling with Gα and the receptor, eventually leading to receptor signaling inhibition. Notably, the desensitization mechanism that involves arrestin deubiquitination is interesting; however, this is a new mechanism and needs to be explored further.
Collapse
|
21
|
Fuentes N, McCullough M, Panettieri RA, Druey KM. RGS proteins, GRKs, and beta-arrestins modulate G protein-mediated signaling pathways in asthma. Pharmacol Ther 2021; 223:107818. [PMID: 33600853 DOI: 10.1016/j.pharmthera.2021.107818] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 12/17/2022]
Abstract
Asthma is a highly prevalent disorder characterized by chronic lung inflammation and reversible airways obstruction. Pathophysiological features of asthma include episodic and reversible airway narrowing due to increased bronchial smooth muscle shortening in response to external and host-derived mediators, excessive mucus secretion into the airway lumen, and airway remodeling. The aberrant airway smooth muscle (ASM) phenotype observed in asthma manifests as increased sensitivity to contractile mediators (EC50) and an increase in the magnitude of contraction (Emax); collectively these attributes have been termed "airways hyper-responsiveness" (AHR). This defining feature of asthma can be promoted by environmental factors including airborne allergens, viruses, and air pollution and other irritants. AHR reduces airway caliber and obstructs airflow, evoking clinical symptoms such as cough, wheezing and shortness of breath. G-protein-coupled receptors (GPCRs) have a central function in asthma through their impact on ASM and airway inflammation. Many but not all treatments for asthma target GPCRs mediating ASM contraction or relaxation. Here we discuss the roles of specific GPCRs, G proteins, and their associated signaling pathways, in asthma, with an emphasis on endogenous mechanisms of GPCR regulation of ASM tone and lung inflammation including regulators of G-protein signaling (RGS) proteins, G-protein coupled receptor kinases (GRKs), and β-arrestin.
Collapse
Affiliation(s)
- Nathalie Fuentes
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States of America
| | - Morgan McCullough
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States of America
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers University School of Medicine, New Brunswick, NJ, United States of America
| | - Kirk M Druey
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States of America.
| |
Collapse
|
22
|
Blankenbach KV, Claas RF, Aster NJ, Spohner AK, Trautmann S, Ferreirós N, Black JL, Tesmer JJG, Offermanns S, Wieland T, Meyer zu Heringdorf D. Dissecting G q/11-Mediated Plasma Membrane Translocation of Sphingosine Kinase-1. Cells 2020; 9:cells9102201. [PMID: 33003441 PMCID: PMC7599897 DOI: 10.3390/cells9102201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/24/2022] Open
Abstract
Diverse extracellular signals induce plasma membrane translocation of sphingosine kinase-1 (SphK1), thereby enabling inside-out signaling of sphingosine-1-phosphate. We have shown before that Gq-coupled receptors and constitutively active Gαq/11 specifically induced a rapid and long-lasting SphK1 translocation, independently of canonical Gq/phospholipase C (PLC) signaling. Here, we further characterized Gq/11 regulation of SphK1. SphK1 translocation by the M3 receptor in HEK-293 cells was delayed by expression of catalytically inactive G-protein-coupled receptor kinase-2, p63Rho guanine nucleotide exchange factor (p63RhoGEF), and catalytically inactive PLCβ3, but accelerated by wild-type PLCβ3 and the PLCδ PH domain. Both wild-type SphK1 and catalytically inactive SphK1-G82D reduced M3 receptor-stimulated inositol phosphate production, suggesting competition at Gαq. Embryonic fibroblasts from Gαq/11 double-deficient mice were used to show that amino acids W263 and T257 of Gαq, which interact directly with PLCβ3 and p63RhoGEF, were important for bradykinin B2 receptor-induced SphK1 translocation. Finally, an AIXXPL motif was identified in vertebrate SphK1 (positions 100–105 in human SphK1a), which resembles the Gαq binding motif, ALXXPI, in PLCβ and p63RhoGEF. After M3 receptor stimulation, SphK1-A100E-I101E and SphK1-P104A-L105A translocated in only 25% and 56% of cells, respectively, and translocation efficiency was significantly reduced. The data suggest that both the AIXXPL motif and currently unknown consequences of PLCβ/PLCδ(PH) expression are important for regulation of SphK1 by Gq/11.
Collapse
Affiliation(s)
- Kira Vanessa Blankenbach
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (K.V.B.); (R.F.C.); (N.J.A.); (A.K.S.)
| | - Ralf Frederik Claas
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (K.V.B.); (R.F.C.); (N.J.A.); (A.K.S.)
| | - Natalie Judith Aster
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (K.V.B.); (R.F.C.); (N.J.A.); (A.K.S.)
| | - Anna Katharina Spohner
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (K.V.B.); (R.F.C.); (N.J.A.); (A.K.S.)
| | - Sandra Trautmann
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (S.T.); (N.F.)
| | - Nerea Ferreirós
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (S.T.); (N.F.)
| | - Justin L. Black
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - John J. G. Tesmer
- Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University West Lafayette, West Lafayette, IN 47907-2054, USA;
| | - Stefan Offermanns
- Abteilung für Pharmakologie, Max-Planck-Institut für Herz- und Lungenforschung, 61231 Bad Nauheim, Germany;
| | - Thomas Wieland
- Experimentelle Pharmakologie Mannheim, European Center for Angioscience, Universität Heidelberg, 68167 Mannheim, Germany;
| | - Dagmar Meyer zu Heringdorf
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (K.V.B.); (R.F.C.); (N.J.A.); (A.K.S.)
- Correspondence: ; Tel.: +49-69-6301-3906
| |
Collapse
|
23
|
Maziarz M, Park JC, Leyme A, Marivin A, Garcia-Lopez A, Patel PP, Garcia-Marcos M. Revealing the Activity of Trimeric G-proteins in Live Cells with a Versatile Biosensor Design. Cell 2020; 182:770-785.e16. [PMID: 32634377 DOI: 10.1016/j.cell.2020.06.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/21/2020] [Accepted: 06/08/2020] [Indexed: 12/28/2022]
Abstract
Heterotrimeric G-proteins (Gαβγ) are the main transducers of signals from GPCRs, mediating the action of countless natural stimuli and therapeutic agents. However, there are currently no robust approaches to directly measure the activity of endogenous G-proteins in cells. Here, we describe a suite of optical biosensors that detect endogenous active G-proteins with sub-second resolution in live cells. Using a modular design principle, we developed genetically encoded, unimolecular biosensors for endogenous Gα-GTP and free Gβγ: the two active species of heterotrimeric G-proteins. This design was leveraged to generate biosensors with specificity for different heterotrimeric G-proteins or for other G-proteins, such as Rho GTPases. Versatility was further validated by implementing the biosensors in multiple contexts, from characterizing cancer-associated G-protein mutants to neurotransmitter signaling in primary neurons. Overall, the versatile biosensor design introduced here enables studying the activity of endogenous G-proteins in live cells with high fidelity, temporal resolution, and convenience.
Collapse
Affiliation(s)
- Marcin Maziarz
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jong-Chan Park
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Anthony Leyme
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Arthur Marivin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Alberto Garcia-Lopez
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Prachi P Patel
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mikel Garcia-Marcos
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
24
|
Zheng M, Zhang X, Sun N, Min X, Acharya S, Kim KM. A novel molecular mechanism responsible for phosphorylation-independent desensitization of G protein-coupled receptors exemplified by the dopamine D 3 receptor. Biochem Biophys Res Commun 2020; 528:432-439. [PMID: 32505358 DOI: 10.1016/j.bbrc.2020.05.197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 05/26/2020] [Indexed: 10/24/2022]
Abstract
GRK-mediated receptor phosphorylation followed by association with β-arrestins has been proposed to be the molecular mechanism involved in the desensitization of G protein-coupled receptors (GPCRs). However, this mechanism does not explain the desensitization of some GPCRs, such as dopamine D3 receptor (D3R), which does not undergo GRK-mediated phosphorylation. Loss-of-function approaches and mutants of dopamine D2 receptor and D3R, which exhibit different desensitization properties, were used to identify the cellular components and processes responsible for desensitization. D3R mediated the recruitment of Mdm2 to the cytosol, which resulted in the constitutive ubiquitination of β-arrestin2 in the resting state. Under desensitization conditions, cytosolic Mdm2 returned to the nucleus, resulting in the deubiquitination of cytosolic β-arrestins. Deubiquitinated β-arrestins formed a tight complex with Gβγ, thereby sequestering it, causing interference in D3R signaling. In conclusion, this study shows that β-arrestins, depending on their ubiquitination status, control the G protein cycling by regulating their interactions with Gβγ. This is a novel mechanism proposed to explain how certain GPCRs can undergo desensitization without receptor phosphorylation.
Collapse
Affiliation(s)
- Mei Zheng
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Xiaohan Zhang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Ningning Sun
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Xiao Min
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Srijan Acharya
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea.
| |
Collapse
|
25
|
Echeverría E, Cabrera M, Burghi V, Sosa M, Ripoll S, Yaneff A, Monczor F, Davio C, Shayo C, Fernández N. The Regulator of G Protein Signaling Homologous Domain of G Protein-Coupled Receptor Kinase 2 Mediates Short-Term Desensitization of β3-Adrenergic Receptor. Front Pharmacol 2020; 11:113. [PMID: 32153413 PMCID: PMC7047201 DOI: 10.3389/fphar.2020.00113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/28/2020] [Indexed: 01/05/2023] Open
Abstract
G protein coupled receptor (GPCR) kinases (GRKs) are key regulators of GPCR signaling. Canonical mechanism of GPCR desensitization involves receptor phosphorylation by GRKs followed by arrestin recruitment and uncoupling from heterotrimeric G protein. Although β3-adrenergic receptor (β3AR) lacks phosphorylation sites by GRKs, agonist treatment proved to induce β3AR desensitization in many cell types. Here we show that GRK2 mediates short-term desensitization of β3AR by a phosphorylation independent mechanism but mediated by its domain homologous to the regulator of G protein signaling (RGS). HEK293T cells overexpressing human β3AR presented a short-term desensitization of cAMP response stimulated by the β3AR agonist, BRL37344, and not by forskolin. We found that β3AR desensitization was higher in cells co-transfected with GRK2. Similarly, overexpression of the RGS homology domain but not kinase domain of GRK2 increased β3AR desensitization. Consistently, stimulation of β3AR increased interaction between GRK2 and Gαs subunit. Furthermore, in rat cardiomyocytes endogenously expressing β3AR, transfection with dominant negative mutant of RH domain of GRK2 (GRK2/D110A) increased cAMP response to BRL37344 and inhibited receptor desensitization. We expect our study to be a starting point for more sophisticated characterization of the consequences of GRK2 mediated desensitization of the β3AR in heart function and disease.
Collapse
Affiliation(s)
- Emiliana Echeverría
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maia Cabrera
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Valeria Burghi
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Máximo Sosa
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sonia Ripoll
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Agustín Yaneff
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Federico Monczor
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos Davio
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carina Shayo
- Laboratorio de Patología y Farmacología Molecular, Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Natalia Fernández
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
26
|
Echeverría E, Velez Rueda AJ, Cabrera M, Juritz E, Burghi V, Fabián L, Davio C, Lorenzano Menna P, Fernández NC. Identification of inhibitors of the RGS homology domain of GRK2 by docking-based virtual screening. Life Sci 2019; 239:116872. [DOI: 10.1016/j.lfs.2019.116872] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 01/25/2023]
|
27
|
Penela P, Ribas C, Sánchez-Madrid F, Mayor F. G protein-coupled receptor kinase 2 (GRK2) as a multifunctional signaling hub. Cell Mol Life Sci 2019; 76:4423-4446. [PMID: 31432234 PMCID: PMC6841920 DOI: 10.1007/s00018-019-03274-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 12/18/2022]
Abstract
Accumulating evidence indicates that G protein-coupled receptor kinase 2 (GRK2) is a versatile protein that acts as a signaling hub by modulating G protein-coupled receptor (GPCR) signaling and also via phosphorylation or scaffolding interactions with an extensive number of non-GPCR cellular partners. GRK2 multifunctionality arises from its multidomain structure and from complex mechanisms of regulation of its expression levels, activity, and localization within the cell, what allows the precise spatio-temporal shaping of GRK2 targets. A better understanding of the GRK2 interactome and its modulation mechanisms is helping to identify the GRK2-interacting proteins and its substrates involved in the participation of this kinase in different cellular processes and pathophysiological contexts.
Collapse
Affiliation(s)
- Petronila Penela
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
| | - Catalina Ribas
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
- Cell-Cell Communication Laboratory, Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | - Federico Mayor
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain.
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain.
| |
Collapse
|
28
|
Abstract
G protein-coupled receptors (GPCRs) are critical cellular sensors that mediate numerous physiological processes. In the heart, multiple GPCRs are expressed on various cell types, where they coordinate to regulate cardiac function by modulating critical processes such as contractility and blood flow. Under pathological settings, these receptors undergo aberrant changes in expression levels, localization and capacity to couple to downstream signalling pathways. Conventional therapies for heart failure work by targeting GPCRs, such as β-adrenergic receptor and angiotensin II receptor antagonists. Although these treatments have improved patient survival, heart failure remains one of the leading causes of mortality worldwide. GPCR kinases (GRKs) are responsible for GPCR phosphorylation and, therefore, desensitization and downregulation of GPCRs. In this Review, we discuss the GPCR signalling pathways and the GRKs involved in the pathophysiology of heart disease. Given that increased expression and activity of GRK2 and GRK5 contribute to the loss of contractile reserve in the stressed and failing heart, inhibition of overactive GRKs has been proposed as a novel therapeutic approach to treat heart failure.
Collapse
|
29
|
Seyedabadi M, Ghahremani MH, Albert PR. Biased signaling of G protein coupled receptors (GPCRs): Molecular determinants of GPCR/transducer selectivity and therapeutic potential. Pharmacol Ther 2019; 200:148-178. [PMID: 31075355 DOI: 10.1016/j.pharmthera.2019.05.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Abstract
G protein coupled receptors (GPCRs) convey signals across membranes via interaction with G proteins. Originally, an individual GPCR was thought to signal through one G protein family, comprising cognate G proteins that mediate canonical receptor signaling. However, several deviations from canonical signaling pathways for GPCRs have been described. It is now clear that GPCRs can engage with multiple G proteins and the line between cognate and non-cognate signaling is increasingly blurred. Furthermore, GPCRs couple to non-G protein transducers, including β-arrestins or other scaffold proteins, to initiate additional signaling cascades. Receptor/transducer selectivity is dictated by agonist-induced receptor conformations as well as by collateral factors. In particular, ligands stabilize distinct receptor conformations to preferentially activate certain pathways, designated 'biased signaling'. In this regard, receptor sequence alignment and mutagenesis have helped to identify key receptor domains for receptor/transducer specificity. Furthermore, molecular structures of GPCRs bound to different ligands or transducers have provided detailed insights into mechanisms of coupling selectivity. However, receptor dimerization, compartmentalization, and trafficking, receptor-transducer-effector stoichiometry, and ligand residence and exposure times can each affect GPCR coupling. Extrinsic factors including cell type or assay conditions can also influence receptor signaling. Understanding these factors may lead to the development of improved biased ligands with the potential to enhance therapeutic benefit, while minimizing adverse effects. In this review, evidence for ligand-specific GPCR signaling toward different transducers or pathways is elaborated. Furthermore, molecular determinants of biased signaling toward these pathways and relevant examples of the potential clinical benefits and pitfalls of biased ligands are discussed.
Collapse
Affiliation(s)
- Mohammad Seyedabadi
- Department of Pharmacology, School of Medicine, Bushehr University of Medical Sciences, Iran; Education Development Center, Bushehr University of Medical Sciences, Iran
| | | | - Paul R Albert
- Ottawa Hospital Research Institute, Neuroscience, University of Ottawa, Canada.
| |
Collapse
|
30
|
Navot S, Kosloff M. Structural design principles that underlie the multi-specific interactions of Gα q with dissimilar partners. Sci Rep 2019; 9:6898. [PMID: 31053791 PMCID: PMC6499889 DOI: 10.1038/s41598-019-43395-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
Gαq is a ubiquitous molecular switch that activates the effectors phospholipase-C-β3 (PLC-β3) and Rho guanine-nucleotide exchange factors. Gαq is inactivated by regulators of G protein signaling proteins, as well as by PLC-β3. Gαq further interacts with G protein-coupled receptor kinase 2 (GRK2), although the functional role of this interaction is debated. While X-ray structures of Gαq bound to representatives of these partners have revealed details of their interactions, the mechanistic basis for differential Gαq interactions with multiple partners (i.e., Gαq multi-specificity) has not been elucidated at the individual residue resolution. Here, we map the structural determinants of Gαq multi-specificity using structure-based energy calculations. We delineate regions that specifically interact with GTPase Activating Proteins (GAPs) and residues that exclusively contribute to effector interactions, showing that only the Gαq “Switch II” region interacts with all partners. Our analysis further suggests that Gαq-GRK2 interactions are consistent with GRK2 functioning as an effector, rather than a GAP. Our multi-specificity analysis pinpoints Gαq residues that uniquely contribute to interactions with particular partners, enabling precise manipulation of these cascades. As such, we dissect the molecular basis of Gαq function as a central signaling hub, which can be used to target Gαq-mediated signaling in therapeutic interventions.
Collapse
Affiliation(s)
- Shir Navot
- The Department of Human Biology, Faculty of Natural Science, University of Haifa, Haifa, 3498838, Israel
| | - Mickey Kosloff
- The Department of Human Biology, Faculty of Natural Science, University of Haifa, Haifa, 3498838, Israel.
| |
Collapse
|
31
|
Murga C, Arcones AC, Cruces-Sande M, Briones AM, Salaices M, Mayor F. G Protein-Coupled Receptor Kinase 2 (GRK2) as a Potential Therapeutic Target in Cardiovascular and Metabolic Diseases. Front Pharmacol 2019; 10:112. [PMID: 30837878 PMCID: PMC6390810 DOI: 10.3389/fphar.2019.00112] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptor kinase 2 (GRK2) is a central signaling node involved in the modulation of many G protein-coupled receptors (GPCRs) and also displaying regulatory functions in other cell signaling routes. GRK2 levels and activity have been reported to be enhanced in patients or in preclinical models of several relevant pathological situations, such as heart failure, cardiac hypertrophy, hypertension, obesity and insulin resistance conditions, or non-alcoholic fatty liver disease (NAFLD), and to contribute to disease progression by a variety of mechanisms related to its multifunctional roles. Therefore, targeting GRK2 by different strategies emerges as a potentially relevant approach to treat cardiovascular disease, obesity, type 2 diabetes, or NAFLD, pathological conditions which are frequently interconnected and present as co-morbidities.
Collapse
Affiliation(s)
- Cristina Murga
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Alba C Arcones
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Marta Cruces-Sande
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Ana M Briones
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Mercedes Salaices
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Federico Mayor
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| |
Collapse
|
32
|
Gurevich VV, Gurevich EV. GPCR Signaling Regulation: The Role of GRKs and Arrestins. Front Pharmacol 2019; 10:125. [PMID: 30837883 PMCID: PMC6389790 DOI: 10.3389/fphar.2019.00125] [Citation(s) in RCA: 341] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/31/2019] [Indexed: 12/13/2022] Open
Abstract
Every animal species expresses hundreds of different G protein-coupled receptors (GPCRs) that respond to a wide variety of external stimuli. GPCRs-driven signaling pathways are involved in pretty much every physiological function and in many pathologies. Therefore, GPCRs are targeted by about a third of clinically used drugs. The signaling of most GPCRs via G proteins is terminated by the phosphorylation of active receptor by specific kinases (GPCR kinases, or GRKs) and subsequent binding of arrestin proteins, that selectively recognize active phosphorylated receptors. In addition, GRKs and arrestins play a role in multiple signaling pathways in the cell, both GPCR-initiated and receptor-independent. Here we focus on the mechanisms of GRK- and arrestin-mediated regulation of GPCR signaling, which includes homologous desensitization and redirection of signaling to additional pathways by bound arrestins.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
33
|
Wolf S, Abd Alla J, Quitterer U. Sensitization of the Angiotensin II AT1 Receptor Contributes to RKIP-Induced Symptoms of Heart Failure. Front Med (Lausanne) 2019; 5:359. [PMID: 30687708 PMCID: PMC6333672 DOI: 10.3389/fmed.2018.00359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/13/2018] [Indexed: 01/30/2023] Open
Abstract
Inhibition of the G-protein-coupled receptor kinase 2 (GRK2) is an emerging treatment approach for heart failure. Therefore, cardio-protective mechanisms induced by GRK2 inhibition are under investigation. We compared two different GRK2 inhibitors, i.e., (i) the dual-specific GRK2 and raf kinase inhibitor protein, RKIP, and (ii) the dominant-negative GRK2-K220R mutant. We found that RKIP induced a strong sensitization of Gq/11-dependent, heart failure-promoting angiotensin II AT1 receptor signaling. The AT1-sensitizing function of RKIP was mediated by the RKIP-GRK2 interaction because the RKIP-S153V mutant, which does not interact with GRK2, had no effect on AT1-stimulated signaling. In contrast, GRK2-K220R significantly inhibited the AT1-stimulated signal. The in vivo relevance of these major differences between two different approaches of GRK2 inhibition was analyzed by generation of transgenic mice with myocardium-specific expression of RKIP and GRK2-K220R. Our results showed that a moderately increased cardiac protein level of RKIP was sufficient to induce major symptoms of heart failure in aged, 8-months-old RKIP-transgenic mice in two different genetic backgrounds. In contrast, GRK2-K220R protected against chronic pressure overload-induced cardiac dysfunction. The AT1 receptor contributed to RKIP-induced heart failure because treatment with the AT1 receptor antagonist, losartan, retarded symptoms of heart failure in RKIP-transgenic mice. Thus, sensitization of the heart failure-promoting AT1 receptor by the RKIP-GRK2 interaction contributes to heart failure whereas dominant-negative GRK2-K220R is cardioprotective. Because RKIP is up-regulated on cardiac biopsy specimens of heart failure patients, the deduced heart failure-promoting mechanism of RKIP could also be relevant for the human disease.
Collapse
Affiliation(s)
- Stefan Wolf
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Joshua Abd Alla
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Ursula Quitterer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, Department of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Maziarz M, Leyme A, Marivin A, Luebbers A, Patel PP, Chen Z, Sprang SR, Garcia-Marcos M. Atypical activation of the G protein Gα q by the oncogenic mutation Q209P. J Biol Chem 2018; 293:19586-19599. [PMID: 30352874 DOI: 10.1074/jbc.ra118.005291] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/15/2018] [Indexed: 12/22/2022] Open
Abstract
The causative role of G protein-coupled receptor (GPCR) pathway mutations in uveal melanoma (UM) has been well-established. Nearly all UMs bear an activating mutation in a GPCR pathway mediated by G proteins of the Gq/11 family, driving tumor initiation and possibly metastatic progression. Thus, targeting this pathway holds therapeutic promise for managing UM. However, direct targeting of oncogenic Gαq/11 mutants, present in ∼90% of UMs, is complicated by the belief that these mutants structurally resemble active Gαq/11 WT. This notion is solidly founded on previous studies characterizing Gα mutants in which a conserved catalytic glutamine (Gln-209 in Gαq) is replaced by leucine, which leads to GTPase function deficiency and constitutive activation. Whereas Q209L accounts for approximately half of GNAQ mutations in UM, Q209P is as frequent as Q209L and also promotes oncogenesis, but has not been characterized at the molecular level. Here, we characterized the biochemical and signaling properties of Gαq Q209P and found that it is also GTPase-deficient and activates downstream signaling as efficiently as Gαq Q209L. However, Gαq Q209P had distinct molecular and functional features, including in the switch II region of Gαq Q209P, which adopted a conformation different from that of Gαq Q209L or active WT Gαq, resulting in altered binding to effectors, Gβγ, and regulators of G-protein signaling (RGS) proteins. Our findings reveal that the molecular properties of Gαq Q209P are fundamentally different from those in other active Gαq proteins and could be leveraged as a specific vulnerability for the ∼20% of UMs bearing this mutation.
Collapse
Affiliation(s)
- Marcin Maziarz
- From the Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Anthony Leyme
- From the Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Arthur Marivin
- From the Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Alex Luebbers
- From the Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Prachi P Patel
- From the Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Zhe Chen
- the Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | - Stephen R Sprang
- the Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana 59812
| | - Mikel Garcia-Marcos
- From the Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118,
| |
Collapse
|
35
|
Abstract
G protein-coupled receptor kinases (GRKs) are classically known for their role in regulating the activity of the largest known class of membrane receptors, which influence diverse biological processes in every cell type in the human body. As researchers have tried to uncover how this family of kinases, containing only 7 members, achieves selective and coordinated control of receptors, they have uncovered a growing number of noncanonical activities for these kinases. These activities include phosphorylation of nonreceptor targets and kinase-independent molecular interactions. In particular, GRK2, GRK3, and GRK5 are the predominant members expressed in the heart. Their canonical and noncanonical actions within cardiac and other tissues have significant implications for cardiovascular function in healthy animals and for the development and progression of disease. This review summarizes what is currently known regarding the activity of these kinases, and particularly the role of GRK2 and GRK5 in the molecular alterations that occur during heart failure. This review further highlights areas of GRK regulation that remain poorly understood and how they may represent novel targets for therapeutic development.
Collapse
|
36
|
Yu S, Sun L, Jiao Y, Lee LTO. The Role of G Protein-coupled Receptor Kinases in Cancer. Int J Biol Sci 2018; 14:189-203. [PMID: 29483837 PMCID: PMC5821040 DOI: 10.7150/ijbs.22896] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/17/2017] [Indexed: 01/14/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of plasma membrane receptors. Emerging evidence demonstrates that signaling through GPCRs affects numerous aspects of cancer biology such as vascular remolding, invasion, and migration. Therefore, development of GPCR-targeted drugs could provide a new therapeutic strategy to treating a variety of cancers. G protein-coupled receptor kinases (GRKs) modulate GPCR signaling by interacting with the ligand-activated GPCR and phosphorylating its intracellular domain. This phosphorylation initiates receptor desensitization and internalization, which inhibits downstream signaling pathways related to cancer progression. GRKs can also regulate non-GPCR substrates, resulting in the modulation of a different set of pathophysiological pathways. In this review, we will discuss the role of GRKs in modulating cell signaling and cancer progression, as well as the therapeutic potential of targeting GRKs.
Collapse
Affiliation(s)
- Shan Yu
- Centre of Reproduction Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Litao Sun
- Department of Ultrasound, The Secondary Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yufei Jiao
- Department of Pathology, The Secondary Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Leo Tsz On Lee
- Centre of Reproduction Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau
| |
Collapse
|
37
|
Signal transduction in L-DOPA-induced dyskinesia: from receptor sensitization to abnormal gene expression. J Neural Transm (Vienna) 2018; 125:1171-1186. [PMID: 29396608 PMCID: PMC6060907 DOI: 10.1007/s00702-018-1847-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/23/2018] [Indexed: 01/06/2023]
Abstract
A large number of signaling abnormalities have been implicated in the emergence and expression of l-DOPA-induced dyskinesia (LID). The primary cause for many of these changes is the development of sensitization at dopamine receptors located on striatal projection neurons (SPN). This initial priming, which is particularly evident at the level of dopamine D1 receptors (D1R), can be viewed as a homeostatic response to dopamine depletion and is further exacerbated by chronic administration of l-DOPA, through a variety of mechanisms affecting various components of the G-protein-coupled receptor machinery. Sensitization of dopamine receptors in combination with pulsatile administration of l-DOPA leads to intermittent and coordinated hyperactivation of signal transduction cascades, ultimately resulting in long-term modifications of gene expression and protein synthesis. A detailed mapping of these pathological changes and of their involvement in LID has been produced during the last decade. According to this emerging picture, activation of sensitized D1R results in the stimulation of cAMP-dependent protein kinase and of the dopamine- and cAMP-regulated phosphoprotein of 32 kDa. This, in turn, activates the extracellular signal-regulated kinases 1 and 2 (ERK), leading to chromatin remodeling and aberrant gene transcription. Dysregulated ERK results also in the stimulation of the mammalian target of rapamycin complex 1, which promotes protein synthesis. Enhanced levels of multiple effector targets, including several transcription factors have been implicated in LID and associated changes in synaptic plasticity and morphology. This article provides an overview of the intracellular modifications occurring in SPN and associated with LID.
Collapse
|
38
|
Nogués L, Palacios-García J, Reglero C, Rivas V, Neves M, Ribas C, Penela P, Mayor F. G protein-coupled receptor kinases (GRKs) in tumorigenesis and cancer progression: GPCR regulators and signaling hubs. Semin Cancer Biol 2018; 48:78-90. [DOI: 10.1016/j.semcancer.2017.04.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/22/2017] [Accepted: 04/26/2017] [Indexed: 12/13/2022]
|
39
|
Yao XQ, Cato MC, Labudde E, Beyett TS, Tesmer JJG, Grant BJ. Navigating the conformational landscape of G protein-coupled receptor kinases during allosteric activation. J Biol Chem 2017; 292:16032-16043. [PMID: 28808053 DOI: 10.1074/jbc.m117.807461] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/10/2017] [Indexed: 01/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are essential for transferring extracellular signals into carefully choreographed intracellular responses controlling diverse aspects of cell physiology. The duration of GPCR-mediated signaling is primarily regulated via GPCR kinase (GRK)-mediated phosphorylation of activated receptors. Although many GRK structures have been reported, the mechanisms underlying GRK activation are not well-understood, in part because it is unknown how these structures map to the conformational landscape available to this enzyme family. Unlike most other AGC kinases, GRKs rely on their interaction with GPCRs for activation and not phosphorylation. Here, we used principal component analysis of available GRK and protein kinase A crystal structures to identify their dominant domain motions and to provide a framework that helps evaluate how close each GRK structure is to being a catalytically competent state. Our results indicated that disruption of an interface formed between the large lobe of the kinase domain and the regulator of G protein signaling homology domain (RHD) is highly correlated with establishment of the active conformation. By introducing point mutations in the GRK5 RHD-kinase domain interface, we show with both in silico and in vitro experiments that perturbation of this interface leads to higher phosphorylation activity. Navigation of the conformational landscape defined by this bioinformatics-based study is likely common to all GPCR-activated GRKs.
Collapse
Affiliation(s)
- Xin-Qiu Yao
- From the Departments of Computational Medicine and Bioinformatics
| | - M Claire Cato
- Biological Chemistry, and.,Life Sciences Institute and
| | | | - Tyler S Beyett
- Life Sciences Institute and.,Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109 and
| | - John J G Tesmer
- Biological Chemistry, and .,Life Sciences Institute and.,Pharmacology, University of Michigan Medical School and
| | - Barry J Grant
- Division of Biological Sciences, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
40
|
Komolov KE, Benovic JL. G protein-coupled receptor kinases: Past, present and future. Cell Signal 2017; 41:17-24. [PMID: 28711719 DOI: 10.1016/j.cellsig.2017.07.004] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 02/08/2023]
Abstract
This review is provided in recognition of the extensive contributions of Dr. Robert J. Lefkowitz to the G protein-coupled receptor (GPCR) field and to celebrate his 75th birthday. Since one of the authors trained with Bob in the 80s, we provide a history of work done in the Lefkowitz lab during the 80s that focused on dissecting the mechanisms that regulate GPCR signaling, with a particular emphasis on the GPCR kinases (GRKs). In addition, we highlight structure/function characteristics of GRK interaction with GPCRs as well as a review of two recent reports that provide a molecular model for GRK-GPCR interaction. Finally, we offer our perspective on some future studies that we believe will drive this field.
Collapse
Affiliation(s)
- Konstantin E Komolov
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States.
| |
Collapse
|
41
|
Steury MD, McCabe LR, Parameswaran N. G Protein-Coupled Receptor Kinases in the Inflammatory Response and Signaling. Adv Immunol 2017; 136:227-277. [PMID: 28950947 DOI: 10.1016/bs.ai.2017.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
G protein-coupled receptor kinases (GRKs) are serine/threonine kinases that regulate a large and diverse class of G protein-coupled receptors (GPCRs). Through GRK phosphorylation and β-arrestin recruitment, GPCRs are desensitized and their signal terminated. Recent work on these kinases has expanded their role from canonical GPCR regulation to include noncanonical regulation of non-GPCR and nonreceptor substrates through phosphorylation as well as via scaffolding functions. Owing to these and other regulatory roles, GRKs have been shown to play a critical role in the outcome of a variety of physiological and pathophysiological processes including chemotaxis, signaling, migration, inflammatory gene expression, etc. This diverse set of functions for these proteins makes them popular targets for therapeutics. Role for these kinases in inflammation and inflammatory disease is an evolving area of research currently pursued in many laboratories. In this review, we describe the current state of knowledge on various GRKs pertaining to their role in inflammation and inflammatory diseases.
Collapse
Affiliation(s)
| | - Laura R McCabe
- Michigan State University, East Lansing, MI, United States
| | | |
Collapse
|
42
|
Lee S, Wottrich S, Bonavida B. Crosstalks between Raf-kinase inhibitor protein and cancer stem cell transcription factors (Oct4, KLF4, Sox2, Nanog). Tumour Biol 2017; 39:1010428317692253. [PMID: 28378634 DOI: 10.1177/1010428317692253] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Raf-kinase inhibitor protein has been reported to inhibit both the Raf/mitogen extracellular signal-regulated kinase/extracellular signal-regulated kinase and nuclear factor kappa-light-chain of activated B cells pathways. It has also been reported in cancers that Raf-kinase inhibitor protein behaves as a metastatic suppressor as well as a chemo-immunosensitizing factor to drug/immune-mediated apoptosis. The majority of cancers exhibit low or no levels of Raf-kinase inhibitor protein. Hence, the activities of Raf-kinase inhibitor protein contrast, in part, to those mediated by several cancer stem cell transcription factors for their roles in resistance and metastasis. In this review, the existence of crosstalks in the signaling pathways between Raf-kinase inhibitor protein and several cancer stem cell transcription factors (Oct4, KLF4, Sox2 and Nanog) was assembled. Oct4 is induced by Lin28, and Raf-kinase inhibitor protein inhibits the microRNA binding protein Lin28. The expression of Raf-kinase inhibitor protein inversely correlates with the expression of Oct4. KLF4 does not interact directly with Raf-kinase inhibitor protein, but rather interacts indirectly via Raf-kinase inhibitor protein's regulation of the Oct4/Sox2/KLF4 complex through the mitogen-activated protein kinase pathway. The mechanism by which Raf-kinase inhibitor protein inhibits Sox2 is via the inhibition of the mitogen-activated protein kinase pathway by Raf-kinase inhibitor protein. Thus, Raf-kinase inhibitor protein's relationship with Sox2 is via its regulation of Oct4. Inhibition of extracellular signal-regulated kinase by Raf-kinase inhibitor protein results in the upregulation of Nanog. The inhibition of Oct4 by Raf-kinase inhibitor protein results in the failure of the heterodimer formation of Oct4 and Sox2 that is necessary to bind to the Nanog promoter for the transcription of Nanog. The findings revealed that there exists a direct correlation between the expression of Raf-kinase inhibitor protein and the expression of each of the above transcription factors. Based on these analyses, we suggest that the expression level of Raf-kinase inhibitor protein may be involved in the regulation of the cancer stem cell phenotype.
Collapse
Affiliation(s)
- SoHyun Lee
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Stephanie Wottrich
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
43
|
Wang J, Luo J, Aryal DK, Wetsel WC, Nass R, Benovic JL. G protein-coupled receptor kinase-2 (GRK-2) regulates serotonin metabolism through the monoamine oxidase AMX-2 in Caenorhabditis elegans. J Biol Chem 2017; 292:5943-5956. [PMID: 28213524 DOI: 10.1074/jbc.m116.760850] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 02/15/2017] [Indexed: 11/06/2022] Open
Abstract
G protein-coupled receptors (GPCRs) regulate many animal behaviors. GPCR signaling is mediated by agonist-promoted interactions of GPCRs with heterotrimeric G proteins, GPCR kinases (GRKs), and arrestins. To further elucidate the role of GRKs in regulating GPCR-mediated behaviors, we utilized the genetic model system Caenorhabditis elegans Our studies demonstrate that grk-2 loss-of-function strains are egg laying-defective and contain low levels of serotonin (5-HT) and high levels of the 5-HT metabolite 5-hydroxyindole acetic acid (5-HIAA). The egg laying defect could be rescued by the expression of wild type but not by catalytically inactive grk-2 or by the selective expression of grk-2 in hermaphrodite-specific neurons. The addition of 5-HT or inhibition of 5-HT metabolism also rescued the egg laying defect. Furthermore, we demonstrate that AMX-2 is the primary monoamine oxidase that metabolizes 5-HT in C. elegans, and we also found that grk-2 loss-of-function strains have abnormally high levels of AMX-2 compared with wild-type nematodes. Interestingly, GRK-2 was also found to interact with and promote the phosphorylation of AMX-2. Additional studies reveal that 5-HIAA functions to inhibit egg laying in a manner dependent on the 5-HT receptor SER-1 and the G protein GOA-1. These results demonstrate that GRK-2 modulates 5-HT metabolism by regulating AMX-2 function and that 5-HIAA may function in the SER-1 signaling pathway.
Collapse
Affiliation(s)
- Jianjun Wang
- From the Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Jiansong Luo
- From the Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | | | - William C Wetsel
- Departments of Psychiatry and Behavioral Sciences.,Cell Biology, and.,Neurobiology and.,Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, North Carolina 27710, and
| | - Richard Nass
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Jeffrey L Benovic
- From the Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107,
| |
Collapse
|
44
|
Dela Paz NG, Melchior B, Frangos JA. Shear stress induces Gα q/11 activation independently of G protein-coupled receptor activation in endothelial cells. Am J Physiol Cell Physiol 2017; 312:C428-C437. [PMID: 28148497 DOI: 10.1152/ajpcell.00148.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 12/17/2022]
Abstract
Mechanochemical signal transduction occurs when mechanical forces, such as fluid shear stress, are converted into biochemical responses within the cell. The molecular mechanisms by which endothelial cells (ECs) sense/transduce shear stress into biological signals, including the nature of the mechanosensor, are still unclear. G proteins and G protein-coupled receptors (GPCRs) have been postulated independently to mediate mechanotransduction. In this study, we used in situ proximity ligation assay (PLA) to investigate the role of a specific GPCR/Gαq/11 pair in EC shear stress-induced mechanotransduction. We demonstrated that sphingosine 1-phosphate (S1P) stimulation causes a rapid dissociation at 0.5 min of Gαq/11 from its receptor S1P3, followed by an increased association within 2 min of GPCR kinase-2 (GRK2) and β-arrestin-1/2 with S1P3 in human coronary artery ECs, which are consistent with GPCR/Gαq/11 activation and receptor desensitization/internalization. The G protein activator AlF4 resulted in increased dissociation of Gαq/11 from S1P3, but no increase in association between S1P3 and either GRK2 or β-arrestin-1/2. The G protein inhibitor guanosine 5'-(β-thio) diphosphate (GDP-β-S) and the S1P3 antagonist VPC23019 both prevented S1P-induced activation. Shear stress also caused the rapid activation within 7 s of S1P3/Gαq/11 There were no increased associations between S1P3 and GRK2 or S1P3 and β-arrestin-1/2 until 5 min. GDP-β-S, but not VPC23019, prevented dissociation of Gαq/11 from S1P3 in response to shear stress. Shear stress did not induce rapid dephosphorylation of β-arrestin-1 or rapid internalization of S1P3, indicating no GPCR activation. These findings suggest that Gαq/11 participates in the sensing/transducing of shear stress independently of GPCR activation in ECs.
Collapse
|
45
|
Rajagopal S, Shenoy SK. GPCR desensitization: Acute and prolonged phases. Cell Signal 2017; 41:9-16. [PMID: 28137506 DOI: 10.1016/j.cellsig.2017.01.024] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 01/25/2017] [Indexed: 01/04/2023]
Abstract
G protein-coupled receptors (GPCRs) transduce a wide array of extracellular signals and regulate virtually every aspect of physiology. While GPCR signaling is essential, overstimulation can be deleterious, resulting in cellular toxicity or uncontrolled cellular growth. Accordingly, nature has developed a number of mechanisms for limiting GPCR signaling, which are broadly referred to as desensitization, and refer to a decrease in response to repeated or continuous stimulation. Short-term desensitization occurs over minutes, and is primarily associated with β-arrestins preventing G protein interaction with a GPCR. Longer-term desensitization, referred to as downregulation, occurs over hours to days, and involves receptor internalization into vesicles, degradation in lysosomes and decreased receptor mRNA levels through unclear mechanisms. Phosphorylation of the receptor by GPCR kinases (GRKs) and the recruitment of β-arrestins is critical to both these short- and long-term desensitization mechanisms. In addition to phosphorylation, both the GPCR and β-arrestins are modified post-translationally in several ways, including by ubiquitination. For many GPCRs, receptor ubiquitination promotes degradation of agonist-activated receptors in the lysosomes. Other proteins also play important roles in desensitization, including phosphodiesterases, RGS family proteins and A-kinase-anchoring proteins. Together, this intricate network of kinases, ubiquitin ligases, and adaptor proteins orchestrate the acute and prolonged desensitization of GPCRs.
Collapse
Affiliation(s)
| | - Sudha K Shenoy
- Department of Medicine (Cardiology), Durham, NC, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
46
|
Gurevich VV, Gurevich EV. Analyzing the roles of multi-functional proteins in cells: The case of arrestins and GRKs. Crit Rev Biochem Mol Biol 2016; 50:440-52. [PMID: 26453028 DOI: 10.3109/10409238.2015.1067185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Most proteins have multiple functions. Obviously, conventional methods of manipulating the level of the protein of interest in the cell, such as over-expression, knockout or knockdown, affect all of its functions simultaneously. The key advantage of these methods is that over-expression, knockout or knockdown does not require any knowledge of the molecular mechanisms of the function(s) of the protein of interest. The disadvantage is that these approaches are inadequate to elucidate the role of an individual function of the protein in a particular cellular process. An alternative is the use of re-engineered proteins, in which a single function is eliminated or enhanced. The use of mono-functional elements of a multi-functional protein can also yield cleaner answers. This approach requires detailed knowledge of the structural basis of each function of the protein in question. Thus, a lot of preliminary structure-function work is necessary to make it possible. However, when this information is available, replacing the protein of interest with a mutant in which individual functions are modified can shed light on the biological role of those particular functions. Here, we illustrate this point using the example of protein kinases, most of which have additional non-enzymatic functions, as well as arrestins, known multi-functional signaling regulators in the cell.
Collapse
Affiliation(s)
| | - Eugenia V Gurevich
- a Department of Pharmacology , Vanderbilt University , Nashville , TN , USA
| |
Collapse
|
47
|
Guccione M, Ettari R, Taliani S, Da Settimo F, Zappalà M, Grasso S. G-Protein-Coupled Receptor Kinase 2 (GRK2) Inhibitors: Current Trends and Future Perspectives. J Med Chem 2016; 59:9277-9294. [PMID: 27362616 DOI: 10.1021/acs.jmedchem.5b01939] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
G-protein-coupled receptor kinase 2 (GRK2) is a G-protein-coupled receptor kinase that is ubiquitously expressed in many tissues and regulates various intracellular mechanisms. The up- or down-regulation of GRK2 correlates with several pathological disorders. GRK2 plays an important role in the maintenance of heart structure and function; thus, this kinase is involved in many cardiovascular diseases. GRK2 up-regulation can worsen cardiac ischemia; furthermore, increased kinase levels occur during the early stages of heart failure and in hypertensive subjects. GRK2 up-regulation can lead to changes in the insulin signaling cascade, which can translate to insulin resistance. Increased GRK2 levels also correlate with the degree of cognitive impairment that is typically observed in Alzheimer's disease. This article reviews the most potent and selective GRK2 inhibitors that have been developed. We focus on their mechanism of action, inhibition profile, and structure-activity relationships to provide insight into the further development of GRK2 inhibitors as drug candidates.
Collapse
Affiliation(s)
- Manuela Guccione
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università degli Studi di Messina , Viale Annunziata, 98168 Messina, Italy
| | - Roberta Ettari
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università degli Studi di Messina , Viale Annunziata, 98168 Messina, Italy
| | - Sabrina Taliani
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno Pisano 6, 56126 Pisa, Italy
| | - Federico Da Settimo
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno Pisano 6, 56126 Pisa, Italy
| | - Maria Zappalà
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università degli Studi di Messina , Viale Annunziata, 98168 Messina, Italy
| | - Silvana Grasso
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università degli Studi di Messina , Viale Annunziata, 98168 Messina, Italy
| |
Collapse
|
48
|
Gurevich EV, Gainetdinov RR, Gurevich VV. G protein-coupled receptor kinases as regulators of dopamine receptor functions. Pharmacol Res 2016; 111:1-16. [PMID: 27178731 DOI: 10.1016/j.phrs.2016.05.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/03/2016] [Accepted: 05/06/2016] [Indexed: 02/08/2023]
Abstract
Actions of the neurotransmitter dopamine in the brain are mediated by dopamine receptors that belong to the superfamily of G protein-coupled receptors (GPCRs). Mammals have five dopamine receptor subtypes, D1 through D5. D1 and D5 couple to Gs/olf and activate adenylyl cyclase, whereas D2, D3, and D4 couple to Gi/o and inhibit it. Most GPCRs upon activation by an agonist are phosphorylated by GPCR kinases (GRKs). The GRK phosphorylation makes receptors high-affinity binding partners for arrestin proteins. Arrestin binding to active phosphorylated receptors stops further G protein activation and promotes receptor internalization, recycling or degradation, thereby regulating their signaling and trafficking. Four non- visual GRKs are expressed in striatal neurons. Here we describe known effects of individual GRKs on dopamine receptors in cell culture and in the two in vivo models of dopamine-mediated signaling: behavioral response to psychostimulants and L-DOPA- induced dyskinesia. Dyskinesia, associated with dopamine super-sensitivity of striatal neurons, is a debilitating side effect of L-DOPA therapy in Parkinson's disease. In vivo, GRK subtypes show greater receptor specificity than in vitro or in cultured cells. Overexpression, knockdown, and knockout of individual GRKs, particularly GRK2 and GRK6, have differential effects on signaling of dopamine receptor subtypes in the brain. Furthermore, deletion of GRK isoforms in select striatal neuronal types differentially affects psychostimulant-induced behaviors. In addition, anti-dyskinetic effect of GRK3 does not require its kinase activity: it is mediated by the binding of its RGS-like domain to Gαq/11, which suppresses Gq/11 signaling. The data demonstrate that the dopamine signaling in defined neuronal types in vivo is regulated by specific and finely orchestrated actions of GRK isoforms.
Collapse
Affiliation(s)
- Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37221, USA.
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia; Skolkovo Institute of Science and Technology, Skolkovo, 143025, Moscow, Russia
| | | |
Collapse
|
49
|
Penela P. Chapter Three - Ubiquitination and Protein Turnover of G-Protein-Coupled Receptor Kinases in GPCR Signaling and Cellular Regulation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:85-140. [PMID: 27378756 DOI: 10.1016/bs.pmbts.2016.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
G-protein-coupled receptors (GPCRs) are responsible for regulating a wide variety of physiological processes, and distinct mechanisms for GPCR inactivation exist to guarantee correct receptor functionality. One of the widely used mechanisms is receptor phosphorylation by specific G-protein-coupled receptor kinases (GRKs), leading to uncoupling from G proteins (desensitization) and receptor internalization. GRKs and β-arrestins also participate in the assembly of receptor-associated multimolecular complexes, thus initiating alternative G-protein-independent signaling events. In addition, the abundant GRK2 kinase has diverse "effector" functions in cellular migration, proliferation, and metabolism homeostasis by means of the phosphorylation or interaction with non-GPCR partners. Altered expression of GRKs (particularly of GRK2 and GRK5) occurs during pathological conditions characterized by impaired GPCR signaling including inflammatory syndromes, cardiovascular disease, and tumor contexts. It is increasingly appreciated that different pathways governing GRK protein stability play a role in the modulation of kinase levels in normal and pathological conditions. Thus, enhanced GRK2 degradation by the proteasome pathway occurs upon GPCR stimulation, what allows cellular adaptation to chronic stimulation in a physiological setting. β-arrestins participate in this process by facilitating GRK2 phosphorylation by different kinases and by recruiting diverse E3 ubiquitin ligase to the receptor complex. Different proteolytic systems (ubiquitin-proteasome, calpains), chaperone activities and signaling pathways influence the stability of GRKs in different ways, thus endowing specificity to GPCR regulation as protein turnover of GRKs can be differentially affected. Therefore, modulation of protein stability of GRKs emerges as a versatile mechanism for feedback regulation of GPCR signaling and basic cellular processes.
Collapse
Affiliation(s)
- P Penela
- Department of Molecular Biology and Centre of Molecular Biology "Severo Ochoa" (CSIC-UAM), Madrid, Autonomous University of Madrid, Madrid, Spain; Spain Health Research Institute The Princesa, Madrid, Spain.
| |
Collapse
|
50
|
Schumacher SM, Gao E, Cohen M, Lieu M, Chuprun JK, Koch WJ. A peptide of the RGS domain of GRK2 binds and inhibits Gα(q) to suppress pathological cardiac hypertrophy and dysfunction. Sci Signal 2016; 9:ra30. [PMID: 27016525 DOI: 10.1126/scisignal.aae0549] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
G protein-coupled receptor (GPCR) kinases (GRKs) play a critical role in cardiac function by regulating GPCR activity. GRK2 suppresses GPCR signaling by phosphorylating and desensitizing active GPCRs, and through protein-protein interactions that uncouple GPCRs from their downstream effectors. Several GRK2 interacting partners, including Gα(q), promote maladaptive cardiac hypertrophy, which leads to heart failure, a leading cause of mortality worldwide. The regulator of G protein signaling (RGS) domain of GRK2 interacts with and inhibits Gα(q) in vitro. We generated TgβARKrgs mice with cardiac-specific expression of the RGS domain of GRK2 and subjected these mice to pressure overload to trigger adaptive changes that lead to heart failure. Unlike their nontransgenic littermate controls, the TgβARKrgs mice exhibited less hypertrophy as indicated by reduced left ventricular wall thickness, decreased expression of genes linked to cardiac hypertrophy, and less adverse structural remodeling. The βARKrgs peptide, but not endogenous GRK2, interacted with Gα(q) and interfered with signaling through this G protein. These data support the development of GRK2-based therapeutic approaches to prevent hypertrophy and heart failure.
Collapse
Affiliation(s)
- Sarah M Schumacher
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA. Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Maya Cohen
- Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Melissa Lieu
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA. Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - J Kurt Chuprun
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA. Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA. Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|