1
|
Egly CL, Barny LA, Do T, McDonald EF, Knollmann BC, Plate L. The proteostasis interactomes of trafficking-deficient variants of the voltage-gated potassium channel K V11.1 associated with long QT syndrome. J Biol Chem 2024; 300:107465. [PMID: 38876300 PMCID: PMC11284683 DOI: 10.1016/j.jbc.2024.107465] [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: 03/28/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024] Open
Abstract
The voltage-gated potassium ion channel KV11.1 plays a critical role in cardiac repolarization. Genetic variants that render Kv11.1 dysfunctional cause long QT syndrome (LQTS), which is associated with fatal arrhythmias. Approximately 90% of LQTS-associated variants cause intracellular protein transport (trafficking) dysfunction, which pharmacological chaperones like E-4031 can rescue. Protein folding and trafficking decisions are regulated by chaperones, protein quality control factors, and trafficking machinery comprising the cellular proteostasis network. Here, we test whether trafficking dysfunction is associated with alterations in the proteostasis network of pathogenic Kv11.1 variants and whether pharmacological chaperones can normalize the proteostasis network of responsive variants. We used affinity-purification coupled with tandem mass tag-based quantitative mass spectrometry to assess protein interaction changes of WT KV11.1 or trafficking-deficient channel variants in the presence or absence of E-4031. We identified 572 core KV11.1 protein interactors. Trafficking-deficient variants KV11.1-G601S and KV11.1-G601S-G965∗ had significantly increased interactions with proteins responsible for folding, trafficking, and degradation compared to WT. We confirmed previous findings that the proteasome is critical for KV11.1 degradation. Our report provides the first comprehensive characterization of protein quality control mechanisms of KV11.1. We find extensive interactome remodeling associated with trafficking-deficient KV11.1 variants and with pharmacological chaperone rescue of KV11.1 cell surface expression. The identified protein interactions could be targeted therapeutically to improve KV11.1 trafficking and treat LQTS.
Collapse
Affiliation(s)
- Christian L Egly
- Department of Medicine, Vanderbilt University Medical Center, Nasville, Tennessee, USA
| | - Lea A Barny
- Program in Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee, USA; Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Tri Do
- Department of Medicine, Vanderbilt University Medical Center, Nasville, Tennessee, USA
| | - Eli F McDonald
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Björn C Knollmann
- Department of Medicine, Vanderbilt University Medical Center, Nasville, Tennessee, USA.
| | - Lars Plate
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
2
|
Egly CL, Barny L, Do T, McDonald EF, Plate L, Knollmann BC. The proteostasis interactomes of trafficking-deficient K V 11.1 variants associated with Long QT Syndrome and pharmacological chaperone rescue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.574410. [PMID: 38352392 PMCID: PMC10862811 DOI: 10.1101/2024.01.31.574410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Introduction The voltage gated potassium ion channel K V 11.1 plays a critical role in cardiac repolarization. Genetic variants that render Kv11.1 dysfunctional cause Long QT Syndrome (LQTS), which is associated with fatal arrhythmias. Approximately 90% of LQTS-associated variants cause intracellular protein transport (trafficking) dysfunction, which can be rescued by pharmacological chaperones like E-4031. Protein folding and trafficking decisions are regulated by chaperones, protein quality control factors, and trafficking machinery, comprising the cellular proteostasis network. Here, we test whether trafficking dysfunction is associated with alterations in the proteostasis network of pathogenic Kv11.1 variants, and whether pharmacological chaperones can normalize the proteostasis network of responsive variants. Methods We used affinity-purification coupled with tandem mass tag-based quantitative mass spectrometry to assess protein interaction changes in human embryonic kidney (HEK293) cells expressing wild-type (WT) K V 11.1 or trafficking-deficient channel variants in the presence or absence of E-4031. Resultsa We identified 573 core K V 11.1 protein interactors. Both variants K V 11.1-G601S and K V 11.1-G601S-G965* had significantly increased interactions with proteins responsible for folding, trafficking, and degradation compared to WT. We found that proteasomal degradation is a key component for K V 11.1 degradation and that the K V 11.1-G601S-G965* variant was more responsive to E-4031 treatment. This suggests a role in the C-terminal domain and the ER retention motif of K V 11.1 in regulating trafficking. Conclusion Our report characterizes the proteostasis network of K V 11.1, two trafficking deficient K V 11.1 variants, and variants treated with a pharmacological chaperone. The identified protein interactions could be targeted therapeutically to improve K V 11.1 trafficking and treat Long QT Syndrome.
Collapse
|
3
|
Grandi E, Navedo MF, Saucerman JJ, Bers DM, Chiamvimonvat N, Dixon RE, Dobrev D, Gomez AM, Harraz OF, Hegyi B, Jones DK, Krogh-Madsen T, Murfee WL, Nystoriak MA, Posnack NG, Ripplinger CM, Veeraraghavan R, Weinberg S. Diversity of cells and signals in the cardiovascular system. J Physiol 2023; 601:2547-2592. [PMID: 36744541 PMCID: PMC10313794 DOI: 10.1113/jp284011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023] Open
Abstract
This white paper is the outcome of the seventh UC Davis Cardiovascular Research Symposium on Systems Approach to Understanding Cardiovascular Disease and Arrhythmia. This biannual meeting aims to bring together leading experts in subfields of cardiovascular biomedicine to focus on topics of importance to the field. The theme of the 2022 Symposium was 'Cell Diversity in the Cardiovascular System, cell-autonomous and cell-cell signalling'. Experts in the field contributed their experimental and mathematical modelling perspectives and discussed emerging questions, controversies, and challenges in examining cell and signal diversity, co-ordination and interrelationships involved in cardiovascular function. This paper originates from the topics of formal presentations and informal discussions from the Symposium, which aimed to develop a holistic view of how the multiple cell types in the cardiovascular system integrate to influence cardiovascular function, disease progression and therapeutic strategies. The first section describes the major cell types (e.g. cardiomyocytes, vascular smooth muscle and endothelial cells, fibroblasts, neurons, immune cells, etc.) and the signals involved in cardiovascular function. The second section emphasizes the complexity at the subcellular, cellular and system levels in the context of cardiovascular development, ageing and disease. Finally, the third section surveys the technological innovations that allow the interrogation of this diversity and advancing our understanding of the integrated cardiovascular function and dysfunction.
Collapse
Affiliation(s)
- Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Donald M. Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Nipavan Chiamvimonvat
- Department of Pharmacology, University of California Davis, Davis, CA, USA
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Rose E. Dixon
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Ana M. Gomez
- Signaling and Cardiovascular Pathophysiology-UMR-S 1180, INSERM, Université Paris-Saclay, Orsay, France
| | - Osama F. Harraz
- Department of Pharmacology, Larner College of Medicine, and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Bence Hegyi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - David K. Jones
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Trine Krogh-Madsen
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, New York, USA
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Matthew A. Nystoriak
- Department of Medicine, Division of Environmental Medicine, Center for Cardiometabolic Science, University of Louisville, Louisville, KY, 40202, USA
| | - Nikki G. Posnack
- Department of Pediatrics, Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
- Sheikh Zayed Institute for Pediatric and Surgical Innovation, Children’s National Heart Institute, Children’s National Hospital, Washington, DC, USA
| | | | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University – Wexner Medical Center, Columbus, OH, USA
| | - Seth Weinberg
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University – Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
4
|
Nguyen NH, Brodsky JL. The cellular pathways that maintain the quality control and transport of diverse potassium channels. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194908. [PMID: 36638864 PMCID: PMC9908860 DOI: 10.1016/j.bbagrm.2023.194908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Potassium channels are multi-subunit transmembrane proteins that permit the selective passage of potassium and play fundamental roles in physiological processes, such as action potentials in the nervous system and organismal salt and water homeostasis, which is mediated by the kidney. Like all ion channels, newly translated potassium channels enter the endoplasmic reticulum (ER) and undergo the error-prone process of acquiring post-translational modifications, folding into their native conformations, assembling with other subunits, and trafficking through the secretory pathway to reach their final destinations, most commonly the plasma membrane. Disruptions in these processes can result in detrimental consequences, including various human diseases. Thus, multiple quality control checkpoints evolved to guide potassium channels through the secretory pathway and clear potentially toxic, aggregation-prone misfolded species. We will summarize current knowledge on the mechanisms underlying potassium channel quality control in the secretory pathway, highlight diseases associated with channel misfolding, and suggest potential therapeutic routes.
Collapse
Affiliation(s)
- Nga H Nguyen
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA 15260, USA.
| |
Collapse
|
5
|
Zhang H, Fu T, Sun J, Zou S, Qiu S, Zhang J, Su S, Shi C, Li DP, Xu Y. Pharmacological suppression of Nedd4-2 rescues the reduction of Kv11.1 channels in pathological cardiac hypertrophy. Front Pharmacol 2022; 13:942769. [PMID: 36059970 PMCID: PMC9428276 DOI: 10.3389/fphar.2022.942769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
The human ether-á-go-go-related gene (hERG) encodes the pore-forming subunit (Kv11.1), conducting a rapidly delayed rectifier K+ current (IKr). Reduction of IKr in pathological cardiac hypertrophy (pCH) contributes to increased susceptibility to arrhythmias. However, practical approaches to prevent IKr deficiency are lacking. Our study investigated the involvement of ubiquitin ligase Nedd4-2-dependent ubiquitination in IKr reduction and sought an intervening approach in pCH. Angiotensin II (Ang II) induced a pCH phenotype in guinea pig, accompanied by increased incidences of sudden death and higher susceptibility to arrhythmias. Patch-clamp recordings revealed a significant IKr reduction in pCH cardiomyocytes. Kv11.1 protein expression was decreased whereas its mRNA level did not change. In addition, Nedd4-2 protein expression was increased in pCH, accompanied by an enhanced Nedd4-2 and Kv11.1 binding detected by immunoprecipitation analysis. Cardiac-specific overexpression of inactive form of Nedd4-2 shortened the prolonged QT interval, reversed IKr reduction, and decreased susceptibility to arrhythmias. A synthesized peptide containing the PY motif in Kv11.1 C-terminus binding to Nedd4-2 and a cell-penetrating sequence antagonized Nedd4-2-dependent degradation of the channel and increased the surface abundance and function of hERG channel in HEK cells. In addition, in vivo administration of the PY peptide shortened QT interval and action potential duration, and enhanced IKr in pCH. We conclude that Nedd4-2-dependent ubiquitination is critically involved in IKr deficiency in pCH. Pharmacological suppression of Nedd4-2 represents a novel approach for antiarrhythmic therapy in pCH.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Tian Fu
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jinglei Sun
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Sihao Zou
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Suhua Qiu
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jiali Zhang
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Shi Su
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Chenxia Shi
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - De-Pei Li
- Center for Precision Medicine, Department of Medicine, School of Medicine University of Missouri, Columbia, MO, United States
- *Correspondence: Yanfang Xu, ; De-Pei Li,
| | - Yanfang Xu
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
- *Correspondence: Yanfang Xu, ; De-Pei Li,
| |
Collapse
|
6
|
Downregulation of hERG Channel Expression By Tyrosine Kinase Inhibitors Nilotinib And Vandetanib Predominantly Contributes To Arrhythmogenesis. Toxicol Lett 2022; 365:11-23. [DOI: 10.1016/j.toxlet.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/11/2022] [Accepted: 06/03/2022] [Indexed: 11/20/2022]
|
7
|
The Role of NEDD4 E3 Ubiquitin–Protein Ligases in Parkinson’s Disease. Genes (Basel) 2022; 13:genes13030513. [PMID: 35328067 PMCID: PMC8950476 DOI: 10.3390/genes13030513] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/03/2022] [Indexed: 01/25/2023] Open
Abstract
Parkinson’s disease (PD) is a debilitating neurodegenerative disease that causes a great clinical burden. However, its exact molecular pathologies are not fully understood. Whilst there are a number of avenues for research into slowing, halting, or reversing PD, one central idea is to enhance the clearance of the proposed aetiological protein, oligomeric α-synuclein. Oligomeric α-synuclein is the main constituent protein in Lewy bodies and neurites and is considered neurotoxic. Multiple E3 ubiquitin-protein ligases, including the NEDD4 (neural precursor cell expressed developmentally downregulated protein 4) family, parkin, SIAH (mammalian homologues of Drosophila seven in absentia), CHIP (carboxy-terminus of Hsc70 interacting protein), and SCFFXBL5 SCF ubiquitin ligase assembled by the S-phase kinase-associated protein (SKP1), cullin-1 (Cul1), a zinc-binding RING finger protein, and the F-box domain/Leucine-rich repeat protein 5-containing protein FBXL5), have been shown to be able to ubiquitinate α-synuclein, influencing its subsequent degradation via the proteasome or lysosome. Here, we explore the link between NEDD4 ligases and PD, which is not only via α-synuclein but further strengthened by several additional substrates and interaction partners. Some members of the NEDD4 family of ligases are thought to crosstalk even with PD-related genes and proteins found to be mutated in familial forms of PD. Mutations in NEDD4 family genes have not been observed in PD patients, most likely because of their essential survival function during development. Following further in vivo studies, it has been thought that NEDD4 ligases may be viable therapeutic targets in PD. NEDD4 family members could clear toxic proteins, enhancing cell survival and slowing disease progression, or might diminish beneficial proteins, reducing cell survival and accelerating disease progression. Here, we review studies to date on the expression and function of NEDD4 ubiquitin ligases in the brain and their possible impact on PD pathology.
Collapse
|
8
|
Ledford HA, Ren L, Thai PN, Park S, Timofeyev V, Sirish P, Xu W, Emigh AM, Priest JR, Perez MV, Ashley EA, Yarov-Yarovoy V, Yamoah EN, Zhang XD, Chiamvimonvat N. Disruption of protein quality control of the human ether-à-go-go related gene K + channel results in profound long QT syndrome. Heart Rhythm 2022; 19:281-292. [PMID: 34634443 PMCID: PMC8810706 DOI: 10.1016/j.hrthm.2021.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 09/20/2021] [Accepted: 10/04/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND Long QT syndrome (LQTS) is a hereditary disease that predisposes patients to life-threatening cardiac arrhythmias and sudden cardiac death. Our previous study of the human ether-à-go-go related gene (hERG)-encoded K+ channel (Kv11.1) supports an association between hERG and RING finger protein 207 (RNF207) variants in aggravating the onset and severity of LQTS, specifically T613M hERG (hERGT613M) and RNF207 frameshift (RNF207G603fs) mutations. However, the underlying mechanistic underpinning remains unknown. OBJECTIVE The purpose of the present study was to test the role of RNF207 in the function of hERG-encoded K+ channel subunits. METHODS Whole-cell patch-clamp experiments were performed in human embryonic kidney (HEK 293) cells and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) together with immunofluorescent confocal and high resolution microscopy, auto-ubiquitinylation assays, and co-immunoprecipitation experiments to test the functional interactions between hERG and RNF207. RESULTS Here, we demonstrated that RNF207 serves as an E3 ubiquitin ligase and targets misfolded hERGT613M proteins for degradation. RNF207G603fs exhibits decreased activity and hinders the normal degradation pathway; this increases the levels of hERGT613M subunits and their dominant-negative effect on the wild-type subunits, ultimately resulting in decreased current density. Similar findings are shown for hERGA614V, a known dominant-negative mutant subunit. Finally, the presence of RNF207G603fs with hERGT613M results in significantly prolonged action potential durations and reduced hERG current in human-induced pluripotent stem cell-derived cardiomyocytes. CONCLUSION Our study establishes RNF207 as an interacting protein serving as a ubiquitin ligase for hERG-encoded K+ channel subunits. Normal function of RNF207 is critical for the quality control of hERG subunits and consequently cardiac repolarization. Moreover, our study provides evidence for protein quality control as a new paradigm in life-threatening cardiac arrhythmias in patients with LQTS.
Collapse
Affiliation(s)
- Hannah A Ledford
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, California
| | - Lu Ren
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, California
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, California
| | - Seojin Park
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, California; Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, Nevada
| | - Valeriy Timofeyev
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, California
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, California; Department of Veterans Affairs, Northern California Health Care System, Mather, California
| | - Wilson Xu
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, California
| | - Aiyana M Emigh
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, California
| | - James R Priest
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California
| | - Marco V Perez
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California
| | - Euan A Ashley
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, California
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, Nevada
| | - Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, California; Department of Veterans Affairs, Northern California Health Care System, Mather, California.
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, California; Department of Veterans Affairs, Northern California Health Care System, Mather, California.
| |
Collapse
|
9
|
Cubeddu LX, de la Rosa D, Ameruoso M. Antiviral and anti-inflammatory drugs to combat COVID-19: Effects on cardiac ion channels and risk of ventricular arrhythmias. BIOIMPACTS : BI 2022; 12:9-20. [PMID: 35087712 PMCID: PMC8783084 DOI: 10.34172/bi.2021.23630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 12/27/2022]
Abstract
![]()
Introduction: Drugs with no indication for the treatment of cardiovascular diseases (e.g., drugs employed to treat COVID-19) can increase the risk of arrhythmias. Of interest, a six-fold increase in the number of arrhythmic events was reported in patients with severe COVID-19. In this study, we reviewed (i) the pro-arrhythmic action of drugs given to patients with COVID-19 infection, and (ii) the effects of inflammatory cytokines on cardiac ion channels and possible generation of arrhythmias.
Methods: We conducted a literature search on the drugs with purported or demonstrated efficacy against COVID-19 disease, emphasizing the mechanisms by which anti-COVID-19 drugs and inflammatory cytokines interfere with cardiac ion channels.
Results:Antibiotics (azithromycin), antimalarials (hydroxychloroquine, chloroquine), antivirals (ritonavir/lopinavir, atazanavir), and some of the tyrosine kinase inhibitors (vandetanib) could induce long QT and increase risk for ventricular arrhythmias. The pro-arrhythmic action results from drug-induced inhibition of Kv11.1 (hERG) channels interfering with the repolarizing potassium IKr currents, leading to long QT and increased risk of triggered arrhythmias. At higher concentrations, these drugs may interfere with IKs, IK1, and/or Ito potassium currents, and even inhibit sodium (INa) and calcium (ICa) currents, inducing additional cardiac toxicity. Ibrutinib, an inhibitor of Bruton’s TK, increased the incidence of atrial fibrillation and ventricular tachycardia associated with a short QT interval. Inflammatory cytokines IL-6 and TNF-α inhibit IKr and Ito repolarizing potassium currents. High levels of inflammatory cytokines could contribute to the arrhythmic events. For remdesivir, favipiravir, dexamethasone, tocilizumab, anakinra, baricitinib, and monoclonal antibodies (bamlanivimab, etesevimab, and casirivimab), no evidence supports significant effects on cardiac ion channels, changes in the QT interval, and increased risk for ventricular arrhythmias.
Conclusion: This study supports the concept of hERG channel promiscuity. Different drug classes given to COVID-19 patients might delay repolarization, and increase the risk of ventricular arrhythmias. The presence of comorbid pro-arrhythmic disease states, and elevated levels of pro-arrhythmic cytokines, could increase the risk of ventricular arrhythmias. Discontinuation of nonessential drugs and correction of electrolyte abnormalities could prevent severe ventricular arrhythmias. Altogether, the most effective therapies against COVID-19 (remdesivir, dexamethasone, monoclonal antibodies) lack pro-arrhythmic activity.
Collapse
Affiliation(s)
- Luigi X Cubeddu
- Nova SE University, Health Professions Division, 3200 S, University Drive, Davie, FL 33328, USA
| | - Daisy de la Rosa
- Nova SE University, Health Professions Division, 3200 S, University Drive, Davie, FL 33328, USA
| | | |
Collapse
|
10
|
The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease. Int J Mol Sci 2021; 22:ijms22116065. [PMID: 34199773 PMCID: PMC8199989 DOI: 10.3390/ijms22116065] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
Despite advances in medicine, cardiac disease remains an increasing health problem associated with a high mortality rate. Maladaptive cardiac remodeling, such as cardiac hypertrophy and fibrosis, is a risk factor for heart failure; therefore, it is critical to identify new therapeutic targets. Failing heart is reported to be associated with hyper-ubiquitylation and impairment of the ubiquitin–proteasome system, indicating an importance of ubiquitylation in the development of cardiac disease. Ubiquitylation is a post-translational modification that plays a pivotal role in protein function and degradation. In 1995, homologous to E6AP C-terminus (HECT) type E3 ligases were discovered. E3 ligases are key enzymes in ubiquitylation and are classified into three families: really interesting new genes (RING), HECT, and RING-between-RINGs (RBRs). Moreover, 28 HECT-type E3 ligases have been identified in human beings. It is well conserved in evolution and is characterized by the direct attachment of ubiquitin to substrates. HECT-type E3 ligase is reported to be involved in a wide range of human diseases and health. The role of HECT-type E3 ligases in the development of cardiac diseases has been uncovered in the last decade. There are only a few review articles summarizing recent advancements regarding HECT-type E3 ligase in the field of cardiac disease. This study focused on cardiac remodeling and described the role of HECT-type E3 ligases in the development of cardiac disease. Moreover, this study revealed that the current knowledge could be exploited for the development of new clinical therapies.
Collapse
|
11
|
Zaza A, Grandi E. Mechanisms of Cav3-associated arrhythmia: Protein or microdomain dysfunction? Int J Cardiol 2020; 320:97-99. [DOI: 10.1016/j.ijcard.2020.06.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 10/23/2022]
|
12
|
Wang Y, Du Y, Luo L, Hu P, Yang G, Li T, Han X, Ma A, Wang T. Alterations of Nedd4-2-binding capacity in PY-motif of Na V 1.5 channel underlie long QT syndrome and Brugada syndrome. Acta Physiol (Oxf) 2020; 229:e13438. [PMID: 31900993 DOI: 10.1111/apha.13438] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/19/2019] [Accepted: 12/29/2019] [Indexed: 12/13/2022]
Abstract
AIMS Pathogenic variants of the SCN5A gene can cause Brugada syndrome (BrS) and long QT syndrome (LQTS), which predispose individuals to potentially fatal ventricular arrhythmias and sudden cardiac death. SCN5A encodes the NaV 1.5 protein, the pore forming α-subunit of the voltage-dependent cardiac Na+ channel. Using a WW domain, the E3 ubiquitin ligase Nedd4-2 binds to the PY-motif ([L/P]PxY) within the C-terminus of NaV 1.5, which results in decreased protein expression and current through NaV 1.5 ubiquitination. Here, we investigate the role of E3 ubiquitin ligase Nedd4-2-mediated NaV 1.5 degradation in the pathological mechanisms of the BrS-associated variant SCN5A-p.L1239P and LQTS-associated variant SCN5A-p.Y1977N. METHODS AND RESULTS Using a combination of molecular biology, biochemical and electrophysiological approaches, we examined the expression, function and Nedd4-2 interactions of SCN5A-p.L1239P and SCN5A-p.Y1977N. SCN5A-p.L1239P is characterized as a loss-of-function, whereas SCN5A-p.Y1977N is a gain-of-function variant of the NaV 1.5 channel. Sequence alignment shows that BrS-associated SCN5A-p.L1239P has a new Nedd4-2-binding site (from LLxY to LPxY). This new Nedd4-2-binding site increases the interaction between NaV 1.5 and Nedd4-2, enhancing ubiquitination and degradation of the NaV 1.5 channel. Disruption of the new Nedd4-2-binding site of SCN5A-p.L1239P restores NaV 1.5 expression and function. However, the LQTS-associated SCN5A-p.Y1977N disrupts the usual Nedd4-2-binding site (from PPxY to PPxN). This decreases NaV 1.5-Nedd4-2 interaction, preventing ubiquitination and degradation of NaV 1.5 channels. CONCLUSIONS Our data suggest that the PY-motif plays an essential role in modifying the expression/function of NaV 1.5 channels through Nedd4-2-mediated ubiquitination. Alterations of NaV 1.5-Nedd4-2 interaction represent a novel pathological mechanism for NaV 1.5 channel diseases caused by SCN5A variants.
Collapse
Affiliation(s)
- Ya Wang
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi P. R. China
| | - Yuan Du
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi P. R. China
| | - Ling Luo
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi P. R. China
| | - Peijing Hu
- Department of Cardiovascular Medicine Second Affiliated Hospital of Xi'an Medical College Xi'an Shaanxi P. R. China
| | - Guodong Yang
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi P. R. China
| | - Tao Li
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi P. R. China
| | - Xiu Han
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi P. R. China
| | - Aiqun Ma
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi P. R. China
- Key Laboratory of Molecular Cardiology Xi'an Shaanxi P. R. China
- Key Laboratory of Environment and Genes Related to Diseases Xi'an Jiaotong University Ministry of Education Xi'an Shaanxi P. R. China
| | - Tingzhong Wang
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi P. R. China
- Key Laboratory of Molecular Cardiology Xi'an Shaanxi P. R. China
- Key Laboratory of Environment and Genes Related to Diseases Xi'an Jiaotong University Ministry of Education Xi'an Shaanxi P. R. China
| |
Collapse
|
13
|
Ye D, Zhou W, Tester DJ, Ackerman MJ. Discovery and characterization of a monogenetic insult, caveolin-3-V37L, that precipitated oligo-proteomic perturbations governing repolarization reserve. Int J Cardiol 2020; 319:71-77. [PMID: 32387251 DOI: 10.1016/j.ijcard.2020.05.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 04/14/2020] [Accepted: 05/04/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Caveolin-3 (Cav-3) is an essential scaffolding protein for caveolae formation in cardiomyocytes and targets multiple long QT syndrome (LQTS)-associated ion channels. Mutations in CAV3 have caused an LQT3-like accentuation in late sodium current, INa (Nav1.5). Here, we characterize a novel CAV3-V37L variant and determine whether it is the substrate for the patient's LQTS. METHODS The proband was a 39-year-old female with drug-induced, sudden cardiac arrest (SCA) with profound QT prolongation (QTc > 600 ms). Genetic testing revealed a rare CAV3-V37L variant of uncertain significance (VUS). Whole-cell patch clamp technique was used to measure IKs, IKr, INa, and ICa, L currents co-expressed with either CAV3-WT or CAV3-V37L in TSA201 cells and to measure the action potential duration (APD) in control human induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) overexpressed with CAV3-WT or CAV3-V37L. RESULTS CAV3-V37L did not affect Nav1.5 late current. Instead, CAV3-V37L resulted in 1) ICa, L with slower inactivation, a 1.5 fold increase in peak ICa, L current density and a 1.1 fold increase in ICa, L persistent current, 2) dramatically reduced IKs peak current density by 74.9%, 3) significantly reduced IKr peak current density by 31.1%, and 4) significantly prolonged the APD in hiPSC-CMs. CONCLUSIONS These functional validation assays enabled the promotion of CAV3-V37L from VUS status to a likely pathogenic variant. Although Nav1.5 was spared, this monogenetic insult precipitated an oligo-proteomic impact with a concomitant gain-of-function of ICa, L and loss-of-function of both IKs and IKr culminating in a marked prolongation of the cardiomyocyte's action potential duration.
Collapse
Affiliation(s)
- Dan Ye
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN 55905, USA; Department of Cardiovascular Medicine/Division of Heart Rhythm Services, Mayo Clinic, Rochester, MN 55905, USA
| | - Wei Zhou
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN 55905, USA; Department of Cardiovascular Medicine/Division of Heart Rhythm Services, Mayo Clinic, Rochester, MN 55905, USA
| | - David J Tester
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN 55905, USA; Department of Cardiovascular Medicine/Division of Heart Rhythm Services, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael J Ackerman
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN 55905, USA; Department of Cardiovascular Medicine/Division of Heart Rhythm Services, Mayo Clinic, Rochester, MN 55905, USA; Department of Pediatric and Adolescent Medicine/Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
14
|
Kim SY, Kim KH, Schilling JM, Leem J, Dhanani M, Head BP, Roth DM, Zemljic-Harpf AE, Patel HH. Protective role of cardiac-specific overexpression of caveolin-3 in cirrhotic cardiomyopathy. Am J Physiol Gastrointest Liver Physiol 2020; 318:G531-G541. [PMID: 31961720 PMCID: PMC7099497 DOI: 10.1152/ajpgi.00346.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cirrhotic cardiomyopathy is a clinical syndrome in patients with liver cirrhosis characterized by blunted cardiac contractile responses to stress and/or heart rate-corrected QT (QTc) interval prolongation. Caveolin-3 (Cav-3) plays a critical role in cardiac protection and is an emerging therapeutic target for heart disease. We investigated the protective role of cardiac-specific overexpression (OE) of Cav-3 in cirrhotic cardiomyopathy. Biliary fibrosis was induced in male Cav-3 OE mice and transgene negative (TGneg) littermates by feeding a diet containing 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC; 0.1%) for 3 wk. Liver pathology and blood chemistries were assessed, and stress echocardiography, telemetry, and isolated heart perfusion studies to assess adrenergic responsiveness were performed. Cav-3 OE mice showed a similar degree of hyperdynamic contractility, pulmonary hypertension, and QTc interval prolongation as TGneg mice after 3 wk of DDC diet. Blunted systolic responses were shown in both DDC-fed Cav-3 OE and TGneg hearts after in vivo isoproterenol challenge. However, QTc interval prolongation after in vivo isoproterenol challenge was significantly less in DDC-fed Cav-3 OE hearts compared with DDC-fed TGneg hearts. In ex vivo perfused hearts, where circulatory factors are absent, isoproterenol challenge showed hearts from DDC-fed Cav-3 OE mice had better cardiac contractility and relaxation compared with DDC-fed TGneg hearts. Although Cav-3 OE in the heart did not prevent cardiac alterations in DDC-induced biliary fibrosis, cardiac expression of Cav-3 reduced QTc interval prolongation after adrenergic stimulation in cirrhosis.NEW & NOTEWORTHY Prevalence of cirrhotic cardiomyopathy is up to 50% in cirrhotic patients, and liver transplantation is the only treatment. However, cirrhotic cardiomyopathy is associated with perioperative morbidity and mortality after liver transplantation; therefore, management of cirrhotic cardiomyopathy is crucial for successful liver transplantation. This study shows cardiac myocyte specific overexpression of caveolin-3 (Cav-3) provides better cardiac contractile responses and less corrected QT prolongation during adrenergic stress in a cirrhotic cardiomyopathy model, suggesting beneficial effects of Cav-3 expression in cirrhotic cardiomyopathy.
Collapse
Affiliation(s)
- So Yeon Kim
- 1Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kang Ho Kim
- 2Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jan M. Schilling
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - Joseph Leem
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - Mehul Dhanani
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - Brian P. Head
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - David M. Roth
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - Alice E. Zemljic-Harpf
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - Hemal H. Patel
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| |
Collapse
|
15
|
Andersson C, Lin H, Liu C, Levy D, Mitchell GF, Larson MG, Vasan RS. Integrated Multiomics Approach to Identify Genetic Underpinnings of Heart Failure and Its Echocardiographic Precursors. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 12:e002489. [DOI: 10.1161/circgen.118.002489] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background:
Heart failure (HF) may arise from alterations in metabolic, structural, and signaling pathways, but its genetic architecture is incompletely understood. To elucidate potential genetic contributors to cardiac remodeling and HF, we integrated genome-wide single-nucleotide polymorphisms, gene expression, and DNA methylation using a transomics analytical approach.
Methods:
We used robust rank aggregation (where the position of a certain gene in a rank order list [based on statistical significance level] is tested against a randomly shuffled rank order list) to derive an integrative transomic score for each annotated gene associated with a HF trait.
Results:
We evaluated ≤8372 FHS (Framingham Heart Study) participants (54% women; mean age, 55±17 years). Of these, 62 (0.7%) and 35 (0.4%) had prevalent HF with reduced ejection fraction and HF with preserved left ventricular ejection fraction, respectively. During a mean follow-up of 8.5 years (minimum–maximum, 0.005–18.6 years), 223 (2.7%) and 234 (2.8%) individuals developed incident HF with reduced ejection fraction and HF with reduced ejection fraction, respectively. Top genes included
MMP20
and
MTSS1
(promotes actin assembly at intercellular junctions) for left ventricular systolic function;
ITGA9
(receptor for
VCAM1
[vascular cell protein 1]) and
C5
for left ventricular remodeling;
NUP210
(expressed during myogenic differentiation) and
ANK1
(cytoskeletal protein) for diastolic function;
TSPAN16
and
RAB11FIP3
(involved in regulation of actin cytoskeleton) for prevalent HF with reduced ejection fraction;
ANKRD13D
and
TRIM69
for incident HF with reduced ejection fraction;
HPCAL1
and
PTTG1IP
for prevalent HF with reduced ejection fraction; and
ZNF146
(close to the
COX7A1
enzyme) and
ZFP3
(close to
SLC52A1
—the riboflavin transporter) for incident HF with reduced ejection fraction. We tested the HF-related top single-nucleotide polymorphisms in the UK biobank, where
rs77059055
in
TPM1
(minor allele frequency, 0.023; odds ratio, 0.83;
P
=0.002) remained statistically significant upon Bonferroni correction.
Conclusions:
Our integrative transomics approach offers insights into potential molecular and genetic contributors to HF and its precursors. Although several of our candidate genes have been implicated in HF in animal models, independent replication is warranted.
Collapse
Affiliation(s)
- Charlotte Andersson
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Department of Cardiology, Herlev and Gentofte Hospital, Herlev, Denmark (C.A.)
| | - Honghuang Lin
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Section of Computational Biomedicine, Department of Medicine (H.L.), Boston University School of Medicine, MA
| | - Chunyu Liu
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Department of Biostatistics (C.L., M.G.L.), Boston University School of Public Health, MA
| | - Daniel Levy
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (D.L.)
| | | | - Martin G. Larson
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Department of Biostatistics (C.L., M.G.L.), Boston University School of Public Health, MA
| | - Ramachandran S. Vasan
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Sections of Preventive Medicine and Epidemiology and Cardiology, Department of Medicine (R.S.V.), Boston University School of Medicine, MA
- Department of Epidemiology (R.S.V.), Boston University School of Public Health, MA
| |
Collapse
|
16
|
Moshal KS, Roder K, Kabakov AY, Werdich AA, Yi-Eng Chiang D, Turan NN, Xie A, Kim TY, Cooper LL, Lu Y, Zhong M, Li W, Terentyev D, Choi BR, Karma A, MacRae CA, Koren G. LITAF (Lipopolysaccharide-Induced Tumor Necrosis Factor) Regulates Cardiac L-Type Calcium Channels by Modulating NEDD (Neural Precursor Cell Expressed Developmentally Downregulated Protein) 4-1 Ubiquitin Ligase. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2019; 12:407-420. [PMID: 31462068 PMCID: PMC6750970 DOI: 10.1161/circgen.119.002641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/19/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The turnover of cardiac ion channels underlying action potential duration is regulated by ubiquitination. Genome-wide association studies of QT interval identified several single-nucleotide polymorphisms located in or near genes involved in protein ubiquitination. A genetic variant upstream of LITAF (lipopolysaccharide-induced tumor necrosis factor) gene prompted us to determine its role in modulating cardiac excitation. METHODS Optical mapping was performed in zebrafish hearts to determine Ca2+ transients. Live-cell confocal calcium imaging was performed on adult rabbit cardiomyocytes to determine intracellular Ca2+handling. L-type calcium channel (LTCC) current (ICa,L) was measured using whole-cell recording. To study the effect of LITAF on Cav1.2 (L-type voltage-gated calcium channel 1.2) channel expression, surface biotinylation, and Westerns were performed. LITAF interactions were studied using coimmunoprecipitation and in situ proximity ligation assay. RESULTS LITAF knockdown in zebrafish resulted in a robust increase in calcium transients. Overexpressed LITAF in 3-week-old rabbit cardiomyocytes resulted in a decrease in ICa,L and Cavα1c abundance, whereas LITAF knockdown increased ICa,L and Cavα1c protein. LITAF-overexpressing decreases calcium transients in adult rabbit cardiomyocytes, which was associated with lower Cavα1c levels. In tsA201 cells, overexpressed LITAF downregulated total and surface pools of Cavα1c via increased Cavα1c ubiquitination and its subsequent lysosomal degradation. We observed colocalization between LITAF and LTCC in tsA201 and cardiomyocytes. In tsA201, NEDD (neural precursor cell expressed developmentally downregulated protein) 4-1, but not its catalytically inactive form NEDD4-1-C867A, increased Cavα1c ubiquitination. Cavα1c ubiquitination was further increased by coexpressed LITAF and NEDD4-1 but not NEDD4-1-C867A. NEDD4-1 knockdown abolished the negative effect of LITAF on ICa,L and Cavα1c levels in 3-week-old rabbit cardiomyocytes. Computer simulations demonstrated that a decrease of ICa,L current associated with LITAF overexpression simultaneously shortened action potential duration and decreased calcium transients in rabbit cardiomyocytes. CONCLUSIONS LITAF acts as an adaptor protein promoting NEDD4-1-mediated ubiquitination and subsequent degradation of LTCC, thereby controlling LTCC membrane levels and function and thus cardiac excitation.
Collapse
Affiliation(s)
- Karni S. Moshal
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Karim Roder
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Anatoli Y. Kabakov
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Andreas A. Werdich
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - David Yi-Eng Chiang
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Nilüfer N. Turan
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - An Xie
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Tae Yun Kim
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | | | - Yichun Lu
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Mingwang Zhong
- Physics Dept & Center for Interdisciplinary Research in Complex Systems, Northeastern Univ, Boston, MA
| | - Weiyan Li
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Dmitry Terentyev
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Bum-Rak Choi
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Alain Karma
- Physics Dept & Center for Interdisciplinary Research in Complex Systems, Northeastern Univ, Boston, MA
| | - Calum A. MacRae
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Gideon Koren
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| |
Collapse
|
17
|
Sutherland-Deveen ME, Wang T, Lamothe SM, Tschirhart JN, Guo J, Li W, Yang T, Du Y, Zhang S. Differential Regulation of Human Ether-à-Go-Go-Related Gene (hERG) Current and Expression by Activation of Protein Kinase C. Mol Pharmacol 2019; 96:1-12. [PMID: 31015282 DOI: 10.1124/mol.118.115188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/17/2019] [Indexed: 02/14/2025] Open
Abstract
The human ether-à-go-go-related gene (hERG) encodes the channel that conducts the rapidly activating delayed rectifier potassium current (IKr) in the heart. Reduction in IKr causes long QT syndrome, which can lead to fatal arrhythmias triggered by stress. One potential link between stress and hERG function is protein kinase C (PKC) activation; however, seemingly conflicting results regarding PKC regulation of hERG have been reported. We investigated the effects of PKC activation using phorbol 12-myristate 13-acetate (PMA) on hERG channels expressed in human embryonic kidney cell line 293 (HEK293) cells and IKr in isolated neonatal rat ventricular myocytes. Acute activation of PKC by PMA (30 nM, 30 minutes) reduced both hERG current (IhERG) and IKr Chronic activation of PKC by PMA (30 nM, 16 hours) increased IKr in cardiomyocytes and the expression level of hERG proteins; however, chronic (30 nM, 16 hours) PMA treatment decreased IhERG, which became larger than untreated control IhERG after PMA removal for 4 hours. Deletion of amino acid residues 2-354 (Δ2-354 hERG) or 1-136 of the N terminus (ΔN 136 hERG) abolished acute PMA (30 nM, 30 minutes)-mediated IhERG reduction. In contrast to wild-type hERG channels, chronic activation of PKC by PMA (30 nM, 16 hours) increased both Δ2-354 hERG and ΔN136 hERG expression levels and currents. The increase in hERG protein was associated with PKC-induced phosphorylation (inhibition) of Nedd4-2, an E3 ubiquitin ligase that mediates hERG degradation. We conclude that PKC regulates hERG in a balanced manner, increasing expression through inhibiting Nedd4-2 while decreasing current through targeting a site(s) within the N terminus.
Collapse
Affiliation(s)
| | - Tingzhong Wang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Shawn M Lamothe
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jared N Tschirhart
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jun Guo
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Wentao Li
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tonghua Yang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Yuan Du
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Shetuan Zhang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
18
|
Mutation-specific peripheral and ER quality control of hERG channel cell-surface expression. Sci Rep 2019; 9:6066. [PMID: 30988392 PMCID: PMC6465299 DOI: 10.1038/s41598-019-42331-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 03/28/2019] [Indexed: 12/11/2022] Open
Abstract
Impaired functional plasma membrane (PM) expression of the hERG K+-channel is associated with Long-QT syndrome type-2 (LQT2) and increased risk of cardiac arrhythmia. Reduced PM-expression is primarily attributed to retention and degradation of misfolded channels by endoplasmic reticulum (ER) protein quality control (QC) systems. However, as the molecular pathogenesis of LQT2 was defined using severely-misfolded hERG variants with limited PM-expression, the potential contribution of post-ER (peripheral) QC pathways to the disease phenotype remains poorly established. Here, we investigate the cellular processing of mildly-misfolded Per-Arnt-Sim (PAS)-domain mutant hERGs, which display incomplete ER-retention and PM-expression defects at physiological temperature. We show that the attenuated PM-expression of hERG is dictated by mutation-specific contributions from both the ER and peripheral QC systems. At the ER, PAS-mutants experience inefficient conformational maturation coupled with rapid ubiquitin-dependent proteasomal degradation. In post-ER compartments, they are rapidly endocytosed from the PM via a ubiquitin-independent mechanism and rapidly targeted for lysosomal degradation. Conformational destabilization underlies aberrant cellular processing at both ER- and post-ER compartments, since conformational correction by a hERG-specific pharmacochaperone or low-temperatures can restore WT-like trafficking. Our results demonstrate that the post-ER QC alone or jointly with the ER QC determines the loss-of-PM-expression phenotype of a subset of LQT2 mutations.
Collapse
|
19
|
Roder K, Kabakov A, Moshal KS, Murphy KR, Xie A, Dudley S, Turan NN, Lu Y, MacRae CA, Koren G. Trafficking of the human ether-a-go-go-related gene (hERG) potassium channel is regulated by the ubiquitin ligase rififylin (RFFL). J Biol Chem 2018; 294:351-360. [PMID: 30401747 DOI: 10.1074/jbc.ra118.003852] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 10/17/2018] [Indexed: 11/06/2022] Open
Abstract
The QT interval is an important diagnostic feature on surface electrocardiograms because it reflects the duration of the ventricular action potential. A previous genome-wide association study has reported a significant linkage between a single-nucleotide polymorphism ∼11.7 kb downstream of the gene encoding the RING finger ubiquitin ligase rififylin (RFFL) and variability in the QT interval. This, along with results in animal studies, suggests that RFFL may have effects on cardiac repolarization. Here, we sought to determine the role of RFFL in cardiac electrophysiology. Adult rabbit cardiomyocytes with adenovirus-expressed RFFL exhibited reduced rapid delayed rectifier current (I Kr). Neonatal rabbit cardiomyocytes transduced with RFFL-expressing adenovirus exhibited reduced total expression of the potassium channel ether-a-go-go-related gene (rbERG). Using transfections of 293A cells and Western blotting experiments, we observed that RFFL and the core-glycosylated form of the human ether-a-go-go-related gene (hERG) potassium channel interact. Furthermore, RFFL overexpression led to increased polyubiquitination and proteasomal degradation of hERG protein and to an almost complete disappearance of I Kr, which depended on the intact RING domain of RFFL. Blocking the ER-associated degradation (ERAD) pathway with a dominant-negative form of the ERAD core component, valosin-containing protein (VCP), in 293A cells partially abolished RFFL-mediated hERG degradation. We further substantiated the link between RFFL and ERAD by showing an interaction between RFFL and VCP in vitro We conclude that RFFL is an important regulator of voltage-gated hERG potassium channel activity and therefore cardiac repolarization and that this ubiquitination-mediated regulation requires parts of the ERAD pathway.
Collapse
Affiliation(s)
- Karim Roder
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903
| | - Anatoli Kabakov
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903
| | - Karni S Moshal
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903
| | - Kevin R Murphy
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903
| | - An Xie
- Department of Medicine, University of Minnesota, Cardiovascular Division, Minneapolis, Minnesota 55455
| | - Samuel Dudley
- Department of Medicine, University of Minnesota, Cardiovascular Division, Minneapolis, Minnesota 55455
| | - Nilüfer N Turan
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903
| | - Yichun Lu
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903
| | - Calum A MacRae
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Gideon Koren
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903.
| |
Collapse
|
20
|
Manning JA, Kumar S. Physiological Functions of Nedd4-2: Lessons from Knockout Mouse Models. Trends Biochem Sci 2018; 43:635-647. [PMID: 30056838 DOI: 10.1016/j.tibs.2018.06.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/11/2018] [Accepted: 06/04/2018] [Indexed: 01/10/2023]
Abstract
Protein modification by ubiquitination plays a key evolutionarily conserved role in regulating membrane proteins. Nedd4-2, a ubiquitin ligase, targets membrane proteins such as ion channels and transporters for ubiquitination. This Nedd4-2-mediated ubiquitination provides a crucial step in controlling the membrane availability of these proteins, thus affecting their signaling and physiological outcomes. In one well-studied example, Nedd4-2 fine-tunes the physiological function of the epithelial sodium channel (ENaC), thus modulating Na+ reabsorption by epithelia to maintain whole-body Na+ homeostasis. This review summarizes the key signaling pathways regulated by Nedd4-2 and the possible implications of such regulation in various pathologies.
Collapse
Affiliation(s)
- Jantina A Manning
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA 5001, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA 5001, Australia.
| |
Collapse
|
21
|
Liu Y, Li D, Nie D, Liu SK, Qiu F, Liu MT, Li YY, Wang JX, Liu YX, Dong CJ, Wu D, Tian W, Yang J, Mu W, Li JT, Zhao D, Wang XF, Chu WF, Yang BF. Arsenic trioxide and angiotensin II have inhibitory effects on HERG protein expression: Evidence for the role of PML SUMOylation. Oncotarget 2018; 8:45447-45458. [PMID: 28525371 PMCID: PMC5542199 DOI: 10.18632/oncotarget.17563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 04/17/2017] [Indexed: 01/30/2023] Open
Abstract
The human ether-a-go-go-related gene (HERG) channel is a novel target for the treatment of drug-induced long QT syndrome, which causes lethal cardiotoxicity. This study is designed to explore the possible role of PML SUMOylation and its associated nuclear bodies (NBs) in the regulation of HERG protein expression. Both arsenic trioxide (ATO) and angiotensin II (Ang II) were able to significantly reduce HERG protein expression, while also increasing PML SUMOylation and accelerating the formation of PML-NBs. Pre-exposure of cardiomyocytes to a SUMOylation chemical inhibitor, ginkgolic acid, or the silencing of UBC9 suppressed PML SUMOylation, subsequently preventing the downregulation of HERG induced by ATO or Ang II. Conversely, knockdown of RNF4 led to a remarkable increase in PML SUMOylation and the function of PML-NBs, further promoting ATO- or Ang II-induced HERG protein downregulation. Mechanistically, an increase in PML SUMOylation by ATO or Ang II dramatically enhanced the formation of PML and Pin1 complexes in PML-NBs, leading to the upregulation of TGF-β1 protein, eventually inhibiting HERG expression through activation of protein kinase A. The present work uncovered a novel molecular mechanism underlying HERG protein expression and indicated that PML SUMOylation is a critical step in the development of drug-acquired arrhythmia.
Collapse
Affiliation(s)
- Yu Liu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Duo Li
- Department of Oral and Maxillofacial Surgery, The 2nd Affiliated Hospital, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Dan Nie
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Shang-Kun Liu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Fang Qiu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Mei-Tong Liu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Yuan-Yuan Li
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Jia-Xin Wang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Yan-Xin Liu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Chang-Jiang Dong
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Di Wu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Wei Tian
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Jia Yang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Wei Mu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Jia-Tong Li
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Dan Zhao
- Department of Clinical Pharmacy, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, The 2nd Affiliated Hospital, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Xiao-Feng Wang
- Department of Oral and Maxillofacial Surgery, The 2nd Affiliated Hospital, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Wen-Feng Chu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| | - Bao-Feng Yang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang 150081, P. R. China
| |
Collapse
|
22
|
Zhu JY, Heidersbach A, Kathiriya IS, Garay BI, Ivey KN, Srivastava D, Han Z, King IN. The E3 ubiquitin ligase Nedd4/Nedd4L is directly regulated by microRNA 1. Development 2017; 144:866-875. [PMID: 28246214 DOI: 10.1242/dev.140368] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 01/10/2017] [Indexed: 12/20/2022]
Abstract
miR-1 is a small noncoding RNA molecule that modulates gene expression in heart and skeletal muscle. Loss of Drosophila miR-1 produces defects in somatic muscle and embryonic heart development, which have been partly attributed to miR-1 directly targeting Delta to decrease Notch signaling. Here, we show that overexpression of miR-1 in the fly wing can paradoxically increase Notch activity independently of its effects on Delta. Analyses of potential miR-1 targets revealed that miR-1 directly regulates the 3'UTR of the E3 ubiquitin ligase Nedd4 Analysis of embryonic and adult fly heart revealed that the Nedd4 protein regulates heart development in Drosophila Larval fly hearts overexpressing miR-1 have profound defects in actin filament organization that are partially rescued by concurrent overexpression of Nedd4. These results indicate that miR-1 and Nedd4 act together in the formation and actin-dependent patterning of the fly heart. Importantly, we have found that the biochemical and genetic relationship between miR-1 and the mammalian ortholog Nedd4-like (Nedd4l) is evolutionarily conserved in the mammalian heart, potentially indicating a role for Nedd4L in mammalian postnatal maturation. Thus, miR-1-mediated regulation of Nedd4/Nedd4L expression may serve to broadly modulate the trafficking or degradation of Nedd4/Nedd4L substrates in the heart.
Collapse
Affiliation(s)
- Jun-Yi Zhu
- Children's National Medical Center, Washington, DC 20010, USA
| | - Amy Heidersbach
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA.,Department of Pediatrics, University of California, San Francisco, CA 94143, USA
| | - Irfan S Kathiriya
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA.,Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Bayardo I Garay
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA.,Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Kathryn N Ivey
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA.,Department of Pediatrics, University of California, San Francisco, CA 94143, USA
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA.,Department of Pediatrics, University of California, San Francisco, CA 94143, USA
| | - Zhe Han
- Children's National Medical Center, Washington, DC 20010, USA
| | - Isabelle N King
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA .,Department of Pediatrics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
23
|
Lamothe SM, Song W, Guo J, Li W, Yang T, Baranchuk A, Graham CH, Zhang S. Hypoxia reduces mature hERG channels through calpain up‐regulation. FASEB J 2017; 31:5068-5077. [DOI: 10.1096/fj.201700255r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/17/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Shawn M. Lamothe
- Department of Biomedical and Molecular SciencesQueen’s UniversityKingstonOntarioCanada
| | - WonJu Song
- Department of Biomedical and Molecular SciencesQueen’s UniversityKingstonOntarioCanada
| | - Jun Guo
- Department of Biomedical and Molecular SciencesQueen’s UniversityKingstonOntarioCanada
| | - Wentao Li
- Department of Biomedical and Molecular SciencesQueen’s UniversityKingstonOntarioCanada
| | - Tonghua Yang
- Department of Biomedical and Molecular SciencesQueen’s UniversityKingstonOntarioCanada
| | - Adrian Baranchuk
- Department of Medicine, Kingston General HospitalQueen’s UniversityKingstonOntarioCanada
| | - Charles H. Graham
- Department of Biomedical and Molecular SciencesQueen’s UniversityKingstonOntarioCanada
| | - Shetuan Zhang
- Department of Biomedical and Molecular SciencesQueen’s UniversityKingstonOntarioCanada
| |
Collapse
|
24
|
Sangoi MG, Lamothe SM, Guo J, Yang T, Li W, Avery EG, Fisher JT, Zhang S. β-Arrestin-Mediated Regulation of the Human Ether-a-go-go-Related Gene Potassium Channel. Mol Pharmacol 2017; 92:162-174. [PMID: 28495999 DOI: 10.1124/mol.116.108035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/10/2017] [Indexed: 02/14/2025] Open
Abstract
The rapidly activating delayed rectifier K+ channel (IKr) is encoded by the human ether-a-go-go-related gene (hERG), which is important for the repolarization of the cardiac action potential. Mutations in hERG or drugs can impair the function or decrease the expression level of hERG channels, leading to long QT syndrome. Thus, it is important to understand hERG channel trafficking and its regulation. For this purpose, G protein-coupled receptors (GPCRs), which regulate a vast array of cellular processes, represent a useful route. The development of designer GPCRs known as designer receptors exclusively activated by designer drugs (DREADDs) has made it possible to dissect specific GPCR signaling pathways in various cellular systems. In the present study, by expressing an arrestin-biased M3 muscarinic receptor-based DREADD (M3D-arr) in stable hERG-expressing human embryonic kidney (HEK) cells, we demonstrate that β-arrestin signaling plays a role in hERG regulation. By exclusively activating M3D-arr using the otherwise inert compound, clozapine-N-oxide, we found that M3D-arr activation increased mature hERG expression and current. Within this paradigm, M3D-arr recruited β-arrestin-1 to the plasma membrane, and promoted phosphoinositide 3-kinase-dependent activation of protein kinase B (Akt). The activated Akt acted through phosphatidylinositol 3-phosphate 5-kinase and Rab11 to facilitate hERG recycling to the plasma membrane. Potential β-arrestin signaling-mediated increases in hERG and IKr were also observed in hERG-HEK cells as well as in neonatal rat ventricular myocytes treated with the muscarinic agonist carbachol. These findings provide novel insight into hERG trafficking and regulation.
Collapse
Affiliation(s)
- Matthew G Sangoi
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Shawn M Lamothe
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jun Guo
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tonghua Yang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Wentao Li
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Ellen G Avery
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - John T Fisher
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Shetuan Zhang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
25
|
Minegishi S, Ishigami T, Kawamura H, Kino T, Chen L, Nakashima-Sasaki R, Doi H, Azushima K, Wakui H, Chiba Y, Tamura K. An Isoform of Nedd4-2 Plays a Pivotal Role in Electrophysiological Cardiac Abnormalities. Int J Mol Sci 2017; 18:ijms18061268. [PMID: 28613240 PMCID: PMC5486090 DOI: 10.3390/ijms18061268] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/12/2017] [Accepted: 06/08/2017] [Indexed: 01/15/2023] Open
Abstract
We have previously shown that neural precursor cell-expressed developmentally downregulated gene 4-2 (Nedd4-2) isoforms with a C2 domain are closely related to ubiquitination of epithelial sodium channel (ENaC), resulting in salt-sensitive hypertension by Nedd4-2 C2 targeting in mice. The sodium voltage-gated channel alpha subunit 5 (SCN5A) gene encodes the α subunit of the human cardiac voltage-gated sodium channel (I Na), and the potassium voltage-gated channel subfamily H member 2 (KCNH2) gene encodes rapidly activating delayed rectifier K channels (I Kr). Both ion channels have also been shown to bind to Nedd4-2 via a conserved Proline-Tyrosine (PY) motif in C-terminal with subsequent ubiquitination and degradation by proteasome. Therefore, loss of Nedd4-2 C2 isoform might be involved in electrophysiological impairment under various conditions. We demonstrate here that Nedd4-2 C2 isoform causes cardiac conduction change in resting condition as well as proarrhythmic change after acute myocardial infarction (MI). The Nedd4-2 C2 knockout (KO) mice showed bradycardia, prolonged QRS, QT intervals, and suppressed PR interval in resting condition. In addition, enhancement of T peak/T end interval was found in mice with surgical ligation of the distal left coronary artery. Morphological analyses based on both ultrasonography of the living heart, as well as histopathological findings revealed that Nedd4-2 C2 KO mice show no significant structural changes from wild-type littermates under resting conditions. These results suggested that Nedd4-2 with C2 domain might play an important role in cardio-renal syndrome through post-transcriptional modification of both ENaC and cardiac ion channels, which are critical for kidney and heart functions.
Collapse
Affiliation(s)
- Shintaro Minegishi
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| | - Tomoaki Ishigami
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| | - Hisho Kawamura
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| | - Tabito Kino
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| | - Lin Chen
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| | - Rie Nakashima-Sasaki
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| | - Hiroshi Doi
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| | - Yumi Chiba
- Department of Nursing, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| |
Collapse
|
26
|
Mahati E, Li P, Kurata Y, Maharani N, Ikeda N, Sakata S, Ogura K, Miake J, Aiba T, Shimizu W, Nakasone N, Ninomiya H, Higaki K, Yamamoto K, Nakai A, Shirayoshi Y, Hisatome I. M3 Muscarinic Receptor Signaling Stabilizes a Novel Mutant Human Ether-a-Go-Go-Related Gene Channel Protein via Phosphorylation of Heat Shock Factor 1 in Transfected Cells. Circ J 2016; 80:2443-2452. [PMID: 27803431 DOI: 10.1253/circj.cj-16-0712] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Long QT syndrome 2 (LQT2) is caused by mutations in the human ether-a-go-go-related gene (hERG). Most of its mutations give rise to unstable hERG proteins degraded by the proteasome. Recently, carbachol was reported to stabilize the wild-type hERG-FLAG via activation of the muscarinic type 3 receptor (M3-mAChR). Its action on mutant hERG-FLAG, however, remains uninvestigated. METHODS AND RESULTS A novel mutant hERG-FLAG carried 2 mutations: an amino acid substitution G572S and an in-frame insertion D1037_V1038insGD. When expressed in HEK293 cells, this mutant hERG-FLAG was degraded by the proteasome and failed to be transported to the cell surface. Carbachol restored stability of the mutant hERG-FLAG and facilitated cell-surface expression. Carbachol activated PKC, augmented phosphorylation of heat shock factor 1 (HSF1) and enhanced expression of heat shock proteins (hsps), hsp70 and hsp90. Both a M3-mAChR antagonist, 4-DAMP, and a PKC inhibitor, bisindolylmaleimide, abolished carbachol-induced stabilization of the mutant hERG-FLAG. CONCLUSIONS M3-mAChR activation leads to enhancement of hsp expression via PKC-dependent phosphorylation of HSF1, thereby stabilizing the mutant hERG-FLAG protein. Thus, M3-mAChR activators may have a therapeutic value for patients with LQT2. (Circ J 2016; 80: 2443-2452).
Collapse
Affiliation(s)
- Endang Mahati
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Spence S, Deurinck M, Ju H, Traebert M, McLean L, Marlowe J, Emotte C, Tritto E, Tseng M, Shultz M, Friedrichs GS. Histone Deacetylase Inhibitors Prolong Cardiac Repolarization through Transcriptional Mechanisms. Toxicol Sci 2016; 153:39-54. [PMID: 27255383 DOI: 10.1093/toxsci/kfw104] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors are an emerging class of anticancer agents that modify gene expression by altering the acetylation status of lysine residues of histone proteins, thereby inducing transcription, cell cycle arrest, differentiation, and cell death or apoptosis of cancer cells. In the clinical setting, treatment with HDAC inhibitors has been associated with delayed cardiac repolarization and in rare instances a lethal ventricular tachyarrhythmia known as torsades de pointes. The mechanism(s) of HDAC inhibitor-induced effects on cardiac repolarization is unknown. We demonstrate that administration of structurally diverse HDAC inhibitors to dogs causes delayed but persistent increases in the heart rate corrected QT interval (QTc), an in vivo measure of cardiac repolarization, at timepoints far removed from the Tmax for parent drug and metabolites. Transcriptional profiling of ventricular myocardium from dogs treated with various HDAC inhibitors demonstrated effects on genes involved in protein trafficking, scaffolding and insertion of various ion channels into the cell membrane as well as genes for specific ion channel subunits involved in cardiac repolarization. Extensive in vitro ion channel profiling of various structural classes of HDAC inhibitors (and their major metabolites) by binding and acute patch clamp assays failed to show any consistent correlations with direct ion channel blockade. Drug-induced rescue of an intracellular trafficking-deficient mutant potassium ion channel, hERG (G601S), and decreased maturation (glycosylation) of wild-type hERG expressed by CHO cells in vitro correlated with prolongation of QTc intervals observed in vivo The results suggest that HDAC inhibitor-induced prolongation of cardiac repolarization may be mediated in part by transcriptional changes of genes required for ion channel trafficking and localization to the sarcolemma. These data have broad implications for the development of these drug classes and suggest that the optimal time to assess potentially transcriptionally mediated physiologic effects will be delayed relative to an epigenetic drug's Tmax/Cmax.
Collapse
Affiliation(s)
- Stan Spence
- *Preclinical Safety, Novartis Institutes of Biomedical Research, 100 Technology Square, Cambridge, MA 02139
| | - Mark Deurinck
- Preclinical Safety, Novartis Institutes of Biomedical Research, Klybeckstrasse 141, CH-4057, Basel, CH
| | - Haisong Ju
- Preclinical Safety, Novartis Institutes of Biomedical Research, One Health Plaza, East Hanover, NJ 07936-1080
| | - Martin Traebert
- Preclinical Safety, Novartis Institutes of Biomedical Research, Klybeckstrasse 141, CH-4057, Basel, CH
| | - LeeAnne McLean
- Oncology Global Development, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ 07936-1080
| | - Jennifer Marlowe
- *Preclinical Safety, Novartis Institutes of Biomedical Research, 100 Technology Square, Cambridge, MA 02139
| | - Corinne Emotte
- Drug Metabolism and Pharmacokinetics, Novartis Pharmaceuticals Corporation, Fabrikstrasse 14-3.02.08, Basel, CH
| | - Elaine Tritto
- Preclinical Safety, Novartis Institutes of Biomedical Research, Klybeckstrasse 141, CH-4057, Basel, CH
| | - Min Tseng
- Development Sciences, Safety Assessment, Investigative Toxicology, Genentech Inc, 550 Grandview Drive, South San Francisco, CA 94080
| | - Michael Shultz
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research Inc, 250 Massachusetts Avenue, Cambridge, MA 02139
| | - Gregory S Friedrichs
- Preclinical Safety, Novartis Institutes of Biomedical Research, One Health Plaza, East Hanover, NJ 07936-1080
| |
Collapse
|
28
|
Foo B, Williamson B, Young JC, Lukacs G, Shrier A. hERG quality control and the long QT syndrome. J Physiol 2016; 594:2469-81. [PMID: 26718903 DOI: 10.1113/jp270531] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/07/2015] [Indexed: 11/08/2022] Open
Abstract
Long-QT syndrome type-2 (LQT2) is characterized by reduced functional expression of the human ether-à-go-go related (hERG) gene product, resulting in impaired cardiac repolarization and predisposition to fatal arrhythmia. Previous studies have implicated abnormal trafficking of misfolded hERG as the primary mechanism of LQT2, with misfolding being caused by mutations in the hERG gene (inherited) or drug treatment (acquired). More generally, environmental and metabolic stresses present a constant challenge to the folding of proteins, including hERG, and must be countered by robust protein quality control (QC) systems. Disposal of partially unfolded yet functional plasma membrane (PM) proteins by protein QC contributes to the loss-of-function phenotype in various conformational diseases including cystic fibrosis (CF) and long-QT syndrome type-2 (LQT2). The prevalent view has been that the loss of PM expression of hERG is attributed to biosynthetic block by endoplasmic reticulum (ER) QC pathways. However, there is a growing appreciation for protein QC pathways acting at post-ER cellular compartments, which may contribute to conformational disease pathogenesis. This article will provide a background on the structure and cellular trafficking of hERG as well as inherited and acquired LQT2. We will review previous work on hERG ER QC and introduce the more novel view that there is a significant peripheral QC at the PM and peripheral cellular compartments. Particular attention is drawn to the unique role of the peripheral QC system in acquired LQT2. Understanding the QC process and players may provide targets for therapeutic intervention in dealing with LQT2.
Collapse
Affiliation(s)
- Brian Foo
- Department of Physiology, McGill University, Montréal, Québec, Canada, H3G 1Y6
| | - Brittany Williamson
- Department of Biochemistry, McGill University, Montréal, Québec, Canada, H3G 1Y6
| | - Jason C Young
- Department of Biochemistry, McGill University, Montréal, Québec, Canada, H3G 1Y6
| | - Gergely Lukacs
- Department of Physiology, McGill University, Montréal, Québec, Canada, H3G 1Y6
| | - Alvin Shrier
- Department of Physiology, McGill University, Montréal, Québec, Canada, H3G 1Y6
| |
Collapse
|
29
|
Chapter Five - Ubiquitination of Ion Channels and Transporters. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:161-223. [DOI: 10.1016/bs.pmbts.2016.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Regulation of the human ether-a-go-go-related gene (hERG) potassium channel by Nedd4 family interacting proteins (Ndfips). Biochem J 2015; 472:71-82. [DOI: 10.1042/bj20141282] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 09/10/2015] [Indexed: 01/01/2023]
Abstract
The human ether-a-go-go-related gene (hERG)-encoded K+ channel is critical for cardiac repolarization. In the present study, we demonstrate that the E3 ubiquitin (Ub) ligase neural precursor cell expressed developmentally down-regulated protein 4-2 (Nedd4-2) is directed to specific cellular compartments by Nedd4 family-interacting proteins (Ndfips) to selectively target the mature hERG channels for degradation.
Collapse
|
31
|
Shi YQ, Yan CC, Zhang X, Yan M, Liu LR, Geng HZ, Lv L, Li BX. Mechanisms underlying probucol-induced hERG-channel deficiency. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3695-704. [PMID: 26229434 PMCID: PMC4516208 DOI: 10.2147/dddt.s86724] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The hERG gene encodes the pore-forming α-subunit of the rapidly activating delayed rectifier potassium channel (I Kr), which is important for cardiac repolarization. Reduction of I hERG due to genetic mutations or drug interferences causes long QT syndrome, leading to life-threatening cardiac arrhythmias (torsades de pointes) or sudden death. Probucol is a cholesterol-lowering drug that could reduce hERG current by decreasing plasma membrane hERG protein expression and eventually cause long QT syndrome. Here, we investigated the mechanisms of probucol effects on I hERG and hERG-channel expression. Our data demonstrated that probucol reduces SGK1 expression, known as SGK isoform, in a concentration-dependent manner, resulting in downregulation of phosphorylated E3 ubiquitin ligase Nedd4-2 expression, but not the total level of Nedd4-2. As a result, the hERG protein reduces, due to the enhanced ubiquitination level. On the contrary, carbachol could enhance the phosphorylation level of Nedd4-2 as an alternative to SGK1, and thus rescue the ubiquitin-mediated degradation of hERG channels caused by probucol. These discoveries provide a novel mechanism of probucol-induced hERG-channel deficiency, and imply that carbachol or its analog may serve as potential therapeutic compounds for the handling of probucol cardiotoxicity.
Collapse
Affiliation(s)
- Yuan-Qi Shi
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Cai-Chuan Yan
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xiao Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Meng Yan
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Li-Rong Liu
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Huai-Ze Geng
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Lin Lv
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Bao-Xin Li
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China ; State-Province Key Laboratory of Biopharmaceutical Engineering, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
32
|
Murfitt L, Whiteley G, Iqbal MM, Kitmitto A. Targeting caveolin-3 for the treatment of diabetic cardiomyopathy. Pharmacol Ther 2015; 151:50-71. [PMID: 25779609 DOI: 10.1016/j.pharmthera.2015.03.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 03/09/2015] [Indexed: 12/21/2022]
Abstract
Diabetes is a global health problem with more than 550 million people predicted to be diabetic by 2030. A major complication of diabetes is cardiovascular disease, which accounts for over two-thirds of mortality and morbidity in diabetic patients. This increased risk has led to the definition of a diabetic cardiomyopathy phenotype characterised by early left ventricular dysfunction with normal ejection fraction. Here we review the aetiology of diabetic cardiomyopathy and explore the involvement of the protein caveolin-3 (Cav3). Cav3 forms part of a complex mechanism regulating insulin signalling and glucose uptake, processes that are impaired in diabetes. Further, Cav3 is key for stabilisation and trafficking of cardiac ion channels to the plasma membrane and so contributes to the cardiac action potential shape and duration. In addition, Cav3 has direct and indirect interactions with proteins involved in excitation-contraction coupling and so has the potential to influence cardiac contractility. Significantly, both impaired contractility and rhythm disturbances are hallmarks of diabetic cardiomyopathy. We review here how changes to Cav3 expression levels and altered relationships with interacting partners may be contributory factors to several of the pathological features identified in diabetic cardiomyopathy. Finally, the review concludes by considering ways in which levels of Cav3 may be manipulated in order to develop novel therapeutic approaches for treating diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Lucy Murfitt
- Institute of Cardiovascular Sciences, Faculty of Medical and Human Sciences, University of Manchester, M13 9NT, UK
| | - Gareth Whiteley
- Institute of Cardiovascular Sciences, Faculty of Medical and Human Sciences, University of Manchester, M13 9NT, UK
| | - Mohammad M Iqbal
- Institute of Cardiovascular Sciences, Faculty of Medical and Human Sciences, University of Manchester, M13 9NT, UK
| | - Ashraf Kitmitto
- Institute of Cardiovascular Sciences, Faculty of Medical and Human Sciences, University of Manchester, M13 9NT, UK.
| |
Collapse
|
33
|
The Cullin 4A/B-DDB1-Cereblon E3 Ubiquitin Ligase Complex Mediates the Degradation of CLC-1 Chloride Channels. Sci Rep 2015; 5:10667. [PMID: 26021757 PMCID: PMC4448132 DOI: 10.1038/srep10667] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 04/22/2015] [Indexed: 12/22/2022] Open
Abstract
Voltage-gated CLC-1 chloride channels play a critical role in controlling the membrane excitability of skeletal muscles. Mutations in human CLC-1 channels have been linked to the hereditary muscle disorder myotonia congenita. We have previously demonstrated that disease-associated CLC-1 A531V mutant protein may fail to pass the endoplasmic reticulum quality control system and display enhanced protein degradation as well as defective membrane trafficking. Currently the molecular basis of protein degradation for CLC-1 channels is virtually unknown. Here we aim to identify the E3 ubiquitin ligase of CLC-1 channels. The protein abundance of CLC-1 was notably enhanced in the presence of MLN4924, a specific inhibitor of cullin-RING E3 ligases. Subsequent investigation with dominant-negative constructs against specific subtypes of cullin-RING E3 ligases suggested that CLC-1 seemed to serve as the substrate for cullin 4A (CUL4A) and 4B (CUL4B). Biochemical examinations further indicated that CUL4A/B, damage-specific DNA binding protein 1 (DDB1), and cereblon (CRBN) appeared to co-exist in the same protein complex with CLC-1. Moreover, suppression of CUL4A/B E3 ligase activity significantly enhanced the functional expression of the A531V mutant. Our data are consistent with the idea that the CUL4A/B-DDB1-CRBN complex catalyses the polyubiquitination and thus controls the degradation of CLC-1 channels.
Collapse
|
34
|
Balycheva M, Faggian G, Glukhov AV, Gorelik J. Microdomain-specific localization of functional ion channels in cardiomyocytes: an emerging concept of local regulation and remodelling. Biophys Rev 2015; 7:43-62. [PMID: 28509981 PMCID: PMC5425752 DOI: 10.1007/s12551-014-0159-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/18/2014] [Indexed: 12/26/2022] Open
Abstract
Cardiac excitation involves the generation of action potential by individual cells and the subsequent conduction of the action potential from cell to cell through intercellular gap junctions. Excitation of the cellular membrane results in opening of the voltage-gated L-type calcium ion (Ca2+) channels, thereby allowing a small amount of Ca2+ to enter the cell, which in turn triggers the release of a much greater amount of Ca2+ from the sarcoplasmic reticulum, the intracellular Ca2+ store, and gives rise to the systolic Ca2+ transient and contraction. These processes are highly regulated by the autonomic nervous system, which ensures the acute and reliable contractile function of the heart and the short-term modulation of this function upon changes in heart rate or workload. It has recently become evident that discrete clusters of different ion channels and regulatory receptors are present in the sarcolemma, where they form an interacting network and work together as a part of a macro-molecular signalling complex which in turn allows the specificity, reliability and accuracy of the autonomic modulation of the excitation-contraction processes by a variety of neurohormonal pathways. Disruption in subcellular targeting of ion channels and associated signalling proteins may contribute to the pathophysiology of a variety of cardiac diseases, including heart failure and certain arrhythmias. Recent methodological advances have made it possible to routinely image the topography of live cardiomyocytes, allowing the study of clustering functional ion channels and receptors as well as their coupling within a specific microdomain. In this review we highlight the emerging understanding of the functionality of distinct subcellular microdomains in cardiac myocytes (e.g. T-tubules, lipid rafts/caveolae, costameres and intercalated discs) and their functional role in the accumulation and regulation of different subcellular populations of sodium, Ca2+ and potassium ion channels and their contributions to cellular signalling and cardiac pathology.
Collapse
Affiliation(s)
- Marina Balycheva
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, 4th Floor National Heart and Lung Institute, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK
- Cardiosurgery Department, University of Verona School of Medicine, Verona, Italy
| | - Giuseppe Faggian
- Cardiosurgery Department, University of Verona School of Medicine, Verona, Italy
| | - Alexey V Glukhov
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, 4th Floor National Heart and Lung Institute, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| | - Julia Gorelik
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, 4th Floor National Heart and Lung Institute, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
35
|
Zhang KP, Yang BF, Li BX. Translational toxicology and rescue strategies of the hERG channel dysfunction: biochemical and molecular mechanistic aspects. Acta Pharmacol Sin 2014; 35:1473-84. [PMID: 25418379 PMCID: PMC4261120 DOI: 10.1038/aps.2014.101] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/20/2014] [Indexed: 01/08/2023]
Abstract
The human ether-à-go-go related gene (hERG) potassium channel is an obligatory anti-target for drug development on account of its essential role in cardiac repolarization and its close association with arrhythmia. Diverse drugs have been removed from the market owing to their inhibitory activity on the hERG channel and their contribution to acquired long QT syndrome (LQTS). Moreover, mutations that cause hERG channel dysfunction may induce congenital LQTS. Recently, an increasing number of biochemical and molecular mechanisms underlying hERG-associated LQTS have been reported. In fact, numerous potential biochemical and molecular rescue strategies are hidden within the biogenesis and regulating network. So far, rescue strategies of hERG channel dysfunction and LQTS mainly include activators, blockers, and molecules that interfere with specific links and other mechanisms. The aim of this review is to discuss the rescue strategies based on hERG channel toxicology from the biochemical and molecular perspectives.
Collapse
Affiliation(s)
- Kai-ping Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, China
- The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China (Key Laboratory of Cardiovascular Research, Ministry of Education), China
| | - Bao-feng Yang
- Department of Pharmacology, Harbin Medical University, Harbin, China
- The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China (Key Laboratory of Cardiovascular Research, Ministry of Education), China
| | - Bao-xin Li
- Department of Pharmacology, Harbin Medical University, Harbin, China
- The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China (Key Laboratory of Cardiovascular Research, Ministry of Education), China
| |
Collapse
|
36
|
Goel P, Manning JA, Kumar S. NEDD4-2 (NEDD4L): the ubiquitin ligase for multiple membrane proteins. Gene 2014; 557:1-10. [PMID: 25433090 DOI: 10.1016/j.gene.2014.11.051] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/14/2014] [Accepted: 11/21/2014] [Indexed: 12/20/2022]
Abstract
NEDD4-2 (also known as NEDD4L, neural precursor cell expressed developmentally down-regulated 4-like) is a ubiquitin protein ligase of the Nedd4 family which is known to bind and regulate a number of membrane proteins to aid in their internalization and turnover. Several of the NEDD4-2 substrates include ion channels, such as the epithelial and voltage-gated sodium channels. Given the critical function of NEDD4-2 in regulating membrane proteins, this ligase is essential for the maintenance of cellular homeostasis. In this article we review the biology and function of this important ubiquitin-protein ligase and discuss its pathophysiological significance.
Collapse
Affiliation(s)
- Pranay Goel
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Jantina A Manning
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, The University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
37
|
Ferreiro SF, Vilariño N, Louzao MC, Nicolaou KC, Frederick MO, Botana LM. In vitro chronic effects on hERG channel caused by the marine biotoxin azaspiracid-2. Toxicon 2014; 91:69-75. [PMID: 25286396 DOI: 10.1016/j.toxicon.2014.09.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/16/2014] [Accepted: 09/25/2014] [Indexed: 11/27/2022]
Abstract
Azaspiracids (AZAs) are marine biotoxins produced by the dinoflagellate Azadinium spinosum that accumulate in many shellfish species. Azaspiracid poisoning caused by AZA-contaminated seafood consumption is primarily manifested by diarrhea in humans. To protect human health, AZA-1, AZA-2 and AZA-3 content in seafood has been regulated by food safety authorities in many countries. Recently AZAs have been reported as a low/moderate hERG channel blockers. Furthermore AZA-2 has been related to arrhythmia appearance in rats, suggesting potential heart toxicity. In this study AZA-2 in vitro effects on hERG channel after chronic exposure are analyzed to further explore potential cardiotoxicity. The amount of hERG channel in the plasma membrane, hERG channel trafficking and hERG currents were evaluated up to 12 h of toxin exposure. In these conditions AZA-2 caused an increase of hERG levels in the plasma membrane, probably related to hERG retrograde trafficking impairment. Although this alteration did not translate into an increase of hERG channel-related current, more studies will be necessary to understand its mechanism and to know what consequences could have in vivo. These findings suggest that azaspiracids might have chronic cardiotoxicity related to hERG channel trafficking and they should not be overlooked when evaluating the threat to human health.
Collapse
Affiliation(s)
- Sara F Ferreiro
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Natalia Vilariño
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| | - M Carmen Louzao
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - K C Nicolaou
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Michael O Frederick
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Luis M Botana
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| |
Collapse
|
38
|
Navarro G, Borroto-Escuela DO, Fuxe K, Franco R. Potential of caveolae in the therapy of cardiovascular and neurological diseases. Front Physiol 2014; 5:370. [PMID: 25324780 PMCID: PMC4179688 DOI: 10.3389/fphys.2014.00370] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/08/2014] [Indexed: 12/25/2022] Open
Abstract
Caveolae are membrane micro-domains enriched in cholesterol, sphingolipids and caveolins, which are transmembrane proteins with a hairpin-like structure. Caveolae participate in receptor-mediated trafficking of cell surface receptors and receptor-mediated signaling. Furthermore, caveolae participate in clathrin-independent endocytosis of membrane receptors. On the one hand, caveolins are involved in vascular and cardiac dysfunction. Also, neurological abnormalities in caveolin-1 knockout mice and a link between caveolin-1 gene haplotypes and neurodegenerative diseases have been reported. The aim of this article is to present the rationale for considering caveolae as potential targets in cardiovascular and neurological diseases.
Collapse
Affiliation(s)
- Gemma Navarro
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona Barcelona, Spain
| | | | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| | - Rafael Franco
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona Barcelona, Spain
| |
Collapse
|
39
|
Wang T, Hogan-Cann A, Kang Y, Cui Z, Guo J, Yang T, Lamothe SM, Li W, Ma A, Fisher JT, Zhang S. Muscarinic receptor activation increases hERG channel expression through phosphorylation of ubiquitin ligase Nedd4-2. Mol Pharmacol 2014; 85:877-86. [PMID: 24688054 DOI: 10.1124/mol.113.091553] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The human ether-à-go-go-related gene (hERG) encodes the pore-forming subunit of the rapidly activating delayed rectifier potassium channel, which is important for cardiac repolarization. Reduction of hERG current due to genetic mutations or drug interferences causes long QT syndrome, leading to cardiac arrhythmias and sudden death. To date, there is no effective therapeutic method to restore or enhance hERG channel function. Using cell biology and electrophysiological methods, we found that the muscarinic receptor agonist carbachol increased the expression and function of hERG, but not ether-à-go-go or Kv1.5 channels stably expressed in human embryonic kidney cells. The carbachol-mediated increase in hERG expression was abolished by the selective M3 antagonist 4-DAMP (1,1-dimethyl-4-diphenylacetoxypiperidinium iodide) but not by the M2 antagonist AF-DX 116 (11[[2-[(diethylamino)methyl]-1-piperidinyl]-acetyl]-5,11-dihydro-6H-pyrido[2,3-b] [1,4]benzodiazepine-6-one). Treatment of cells with carbachol reduced the hERG-ubiquitin interaction and slowed the rate of hERG degradation. We previously showed that the E3 ubiquitin ligase Nedd4-2 mediates degradation of hERG channels. Here, we found that disrupting the Nedd4-2 binding domain in hERG completely eliminated the effect of carbachol on hERG channels. Carbachol treatment enhanced the phosphorylation level, but not the total level, of Nedd4-2. Blockade of the protein kinase C (PKC) pathway abolished the carbachol-induced enhancement of hERG channels. Our data suggest that muscarinic activation increases hERG channel expression by phosphorylating Nedd4-2 via the PKC pathway.
Collapse
Affiliation(s)
- Tingzhong Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China (T.W., A.M.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (T.W., A.H.-C., Y.K., Z.C., J.G., T.Y., S.M.L., W.L., J.T.F., S.Z.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Mehta A, Sequiera GL, Ramachandra CJA, Sudibyo Y, Chung Y, Sheng J, Wong KY, Tan TH, Wong P, Liew R, Shim W. Re-trafficking of hERG reverses long QT syndrome 2 phenotype in human iPS-derived cardiomyocytes. Cardiovasc Res 2014; 102:497-506. [PMID: 24623279 DOI: 10.1093/cvr/cvu060] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIMS Long QT syndrome 2 (LQTS2) caused by missense mutations in hERG channel is clinically associated with abnormally prolonged ventricular repolarization and sudden cardiac deaths. Modelling monogenic arrhythmogenic diseases using human-induced pluripotent stem cells (hiPSCs) offers unprecedented mechanistic insights into disease pathogenesis. We utilized LQTS2-hiPSC-derived cardiomyocytes (CMs) to elucidate pathological changes and to demonstrate reversal of LQTS2 phenotype in a therapeutic intervention using a pharmacological agent, (N-[N-(N-acetyl-l-leucyl)-l-leucyl]-l-norleucine) (ALLN). METHODS AND RESULTS We generated LQTS2-specific CMs (A561V missense mutation in KCNH2) from iPSCs using the virus-free reprogramming method. These CMs recapitulate dysfunction of hERG potassium channel with diminished IKr currents, prolonged repolarization durations, and elevated arrhythmogenesis due to reduced membrane localization of glycosylated/mature hERG. Dysregulated expression of folding chaperones and processing proteasomes coupled with sequestered hERG in the endoplasmic reticulum confirmed trafficking-induced disease manifestation. Treatment with ALLN, not only increased membrane localization of mature hERG but also reduced repolarization, increased IKr currents and reduced arrhythmogenic events. Diverged from biophysical interference of hERG channel, our results show that modulation of chaperone proteins could be therapeutic in LQTS2 treatment. CONCLUSION Our in vitro study shows an alternative approach to rescue diseased LQTS2 phenotype via corrective re-trafficking therapy using a small chemical molecule, such as ALLN. This potentially novel approach may have ramifications in other clinically relevant trafficking disorders.
Collapse
Affiliation(s)
- Ashish Mehta
- Research and Development Unit, National Heart Centre Singapore, Singapore National Heart Research Institute Singapore, Singapore
| | | | | | - Yuliansa Sudibyo
- Research and Development Unit, National Heart Centre Singapore, Singapore
| | - Yingying Chung
- Research and Development Unit, National Heart Centre Singapore, Singapore
| | - Jingwei Sheng
- Research and Development Unit, National Heart Centre Singapore, Singapore
| | - Keng Yean Wong
- Department of Paediatrics, KK Women's and Children Hospital, Singapore
| | - Teng Hong Tan
- Department of Paediatrics, KK Women's and Children Hospital, Singapore
| | - Philip Wong
- Department of Cardiology, National Heart Centre Singapore, Singapore Cardiovascular and Metabolic Disorders Program, DUKE-NUS, Singapore
| | - Reginald Liew
- Department of Cardiology, National Heart Centre Singapore, Singapore Cardiovascular and Metabolic Disorders Program, DUKE-NUS, Singapore
| | - Winston Shim
- Research and Development Unit, National Heart Centre Singapore, Singapore National Heart Research Institute Singapore, Singapore Cardiovascular and Metabolic Disorders Program, DUKE-NUS, Singapore
| |
Collapse
|
41
|
Pakladok T, Hosseinzadeh Z, Almilaji A, Lebedeva A, Shumilina E, Alesutan I, Lang F. Up-regulation of hERG K⁺ channels by B-RAF. PLoS One 2014; 9:e87457. [PMID: 24475291 PMCID: PMC3903650 DOI: 10.1371/journal.pone.0087457] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 12/26/2013] [Indexed: 11/19/2022] Open
Abstract
Human ether-a-go-go related-gene K⁺ channels (hERG) participate in the regulation of tumor cell proliferation and apoptosis. HERG channel activity is up-regulated by growth factors. Kinases sensitive to growth factor signaling include the serine/threonine protein kinase B-RAF. The present study thus explored whether B-RAF influences hERG channel expression and activity. To this end, hERG channels were expressed in Xenopus oocytes with or without wild-type B-RAF, hERG channel activity was determined utilizing dual-electrode voltage clamp and hERG protein abundance in the cell membrane was analyzed utilizing confocal microscopy as well as chemiluminescence. Moreover, in rhabdomyosarcoma RD cells the effect of B-RAF inhibitor PLX-4720 on hERG-mediated current was quantified by whole-cell patch clamp and hERG cell surface protein abundance by utilizing biotinylation of cell surface proteins as well as flow cytometry. As a result, co-expression of wild-type B-RAF in hERG-expressing Xenopus oocytes significantly increased hERG channel activity and hERG channel protein abundance in the cell membrane. Treatment for 24 hours of B-RAF and hERG-expressing Xenopus oocytes with B-RAF inhibitor PLX-4720 (10 µM) significantly decreased hERG-mediated current and hERG cell surface expression. Similarly, in rhabdomyosarcoma RD cells, treatment for 24 hours with B-RAF inhibitor PLX-4720 significantly decreased hERG cell membrane protein abundance and hERG-mediated current. In conclusion, B-RAF is a powerful regulator of hERG channel activity and cell surface hERG protein abundance.
Collapse
Affiliation(s)
| | | | - Ahmad Almilaji
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Aleksandra Lebedeva
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Immunology, Institute of Experimental Medicine, St. Petersburg, Russia
| | | | - Ioana Alesutan
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
42
|
Wilkars W, Wollberg J, Mohr E, Han M, Chetkovich DM, Bähring R, Bender RA. Nedd4‐2 regulates surface expression and may affect
N
‐glycosylation of hyperpolarization‐activated cyclic nucleotide‐gated (HCN)‐1 channels. FASEB J 2014; 28:2177-90. [DOI: 10.1096/fj.13-242032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Wiebke Wilkars
- Institute of NeuroanatomyUniversity of Hamburg Medical CenterHamburgGermany
| | - Jessica Wollberg
- Institute of Cellular and Integrative PhysiologyUniversity of Hamburg Medical CenterHamburgGermany
| | - Evita Mohr
- Institute of NeuroanatomyUniversity of Hamburg Medical CenterHamburgGermany
| | - Mieri Han
- Institute of NeuroanatomyUniversity of Hamburg Medical CenterHamburgGermany
| | - Dane M. Chetkovich
- Davee Department of Neurology and Clinical NeurosciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of PhysiologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Robert Bähring
- Institute of Cellular and Integrative PhysiologyUniversity of Hamburg Medical CenterHamburgGermany
| | - Roland A. Bender
- Institute of NeuroanatomyUniversity of Hamburg Medical CenterHamburgGermany
| |
Collapse
|
43
|
An H, Krist DT, Statsyuk AV. Crosstalk between kinases and Nedd4 family ubiquitin ligases. ACTA ACUST UNITED AC 2014; 10:1643-57. [DOI: 10.1039/c3mb70572b] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Understanding the interplay between kinase and E3 ligase signaling pathways will allow better understanding of therapeutically relevant pathways and the design of small molecule therapeutics targeting these pathways.
Collapse
Affiliation(s)
- Heeseon An
- Department of Chemistry
- Northwestern University
- Evanston, USA
| | - David T. Krist
- Department of Chemistry
- Northwestern University
- Evanston, USA
| | | |
Collapse
|
44
|
Willis MS, Bevilacqua A, Pulinilkunnil T, Kienesberger P, Tannu M, Patterson C. The role of ubiquitin ligases in cardiac disease. J Mol Cell Cardiol 2013; 71:43-53. [PMID: 24262338 DOI: 10.1016/j.yjmcc.2013.11.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 11/08/2013] [Accepted: 11/11/2013] [Indexed: 01/13/2023]
Abstract
Rigorous surveillance of protein quality control is essential for the maintenance of normal cardiac function, while the dysregulation of protein turnover is present in a diverse array of common cardiac diseases. Central to the protein quality control found in all cells is the ubiquitin proteasome system (UPS). The UPS plays a critical role in protein trafficking, cellular signaling, and most prominently, protein degradation. As ubiquitin ligases (E3s) control the specificity of the UPS, their description in the cardiomyocyte has highlighted how ubiquitin ligases are critical to the turnover and function of the sarcomere complex, responsible for the heart's required continuous contraction. In this review, we provide an overview of the UPS, highlighting a comprehensive overview of the cardiac ubiquitin ligases identified to date. We then focus on recent studies of new cardiac ubiquitin ligases outlining their novel roles in protein turnover, cellular signaling, and the regulation of mitochondrial dynamics and receptor turnover in the pathophysiology of cardiac hypertrophy, cardiac atrophy, myocardial infarction, and heart failure. This article is part of a Special Issue entitled "Protein Quality Control, the Ubiquitin Proteasome System, and Autophagy".
Collapse
Affiliation(s)
- Monte S Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.
| | - Ariana Bevilacqua
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Dalhousie University, Saint John, NB, Canada
| | - Petra Kienesberger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Saint John, NB, Canada
| | - Manasi Tannu
- College of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cam Patterson
- Departments of Cell and Developmental Biology, Medicine (Cardiology), and Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
45
|
Cutler MJ, Kaufman ES. To be or not to be: long-QT syndrome type 9. CIRCULATION. CARDIOVASCULAR GENETICS 2013; 6:439-440. [PMID: 24129590 DOI: 10.1161/circgenetics.113.000345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Affiliation(s)
- Michael J Cutler
- Heart & Vascular Center, MetroHealth Campus of Case Western Reserve University, Cleveland, OH
| | | |
Collapse
|
46
|
Mulet JM, Llopis-Torregrosa V, Primo C, Marqués MC, Yenush L. Endocytic regulation of alkali metal transport proteins in mammals, yeast and plants. Curr Genet 2013; 59:207-30. [PMID: 23974285 DOI: 10.1007/s00294-013-0401-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/24/2013] [Accepted: 07/29/2013] [Indexed: 12/30/2022]
Abstract
The relative concentrations of ions and solutes inside cells are actively maintained by several classes of transport proteins, in many cases against their concentration gradient. These transport processes, which consume a large portion of cellular energy, must be constantly regulated. Many structurally distinct families of channels, carriers, and pumps have been characterized in considerable detail during the past decades and defects in the function of some of these proteins have been linked to a growing list of human diseases. The dynamic regulation of the transport proteins present at the cell surface is vital for both normal cellular function and for the successful adaptation to changing environments. The composition of proteins present at the cell surface is controlled on both the transcriptional and post-translational level. Post-translational regulation involves highly conserved mechanisms of phosphorylation- and ubiquitylation-dependent signal transduction routes used to modify the cohort of receptors and transport proteins present under any given circumstances. In this review, we will summarize what is currently known about one facet of this regulatory process: the endocytic regulation of alkali metal transport proteins. The physiological relevance, major contributors, parallels and missing pieces of the puzzle in mammals, yeast and plants will be discussed.
Collapse
Affiliation(s)
- José Miguel Mulet
- Instituto de Biología Molecular y Celular de Plantas (IBMCP), Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, Avd. de los Naranjos s/n, 46022, Valencia, Spain
| | | | | | | | | |
Collapse
|
47
|
Cui Z, Zhang S. Regulation of the human ether-a-go-go-related gene (hERG) channel by Rab4 protein through neural precursor cell-expressed developmentally down-regulated protein 4-2 (Nedd4-2). J Biol Chem 2013; 288:21876-86. [PMID: 23792956 DOI: 10.1074/jbc.m113.461715] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The human ether-a-go-go-related gene (hERG) encodes the pore-forming α-subunit of the rapidly activating delayed rectifier K(+) channel in the heart, which plays a critical role in cardiac action potential repolarization. Dysfunction of IKr causes long QT syndrome, a cardiac electrical disorder that predisposes affected individuals to fatal arrhythmias and sudden death. The homeostasis of hERG channels in the plasma membrane depends on a balance between protein synthesis and degradation. Our recent data indicate that hERG channels undergo enhanced endocytic degradation under low potassium (hypokalemia) conditions. The GTPase Rab4 is known to mediate rapid recycling of various internalized proteins to the plasma membrane. In the present study, we investigated the effect of Rab4 on the expression level of hERG channels. Our data revealed that overexpression of Rab4 decreases the expression level of hERG in the plasma membrane. Rab4 does not affect the expression level of the Kv1.5 or EAG K(+) channels. Mechanistically, our data demonstrate that overexpression of Rab4 increases the expression level of endogenous Nedd4-2, a ubiquitin ligase that targets hERG but not Kv1.5 or EAG channels for ubiquitination and degradation. Nedd4-2 undergoes self- ubiquitination and degradation. Rab4 interferes with Nedd4-2 degradation, resulting in an increased expression level of Nedd4-2, which targets hERG. In summary, the present study demonstrates a novel pathway for hERG regulation; Rab4 decreases the hERG density at the plasma membrane by increasing the endogenous Nedd4-2 expression.
Collapse
Affiliation(s)
- Zhi Cui
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | | |
Collapse
|
48
|
Almilaji A, Munoz C, Elvira B, Fajol A, Pakladok T, Honisch S, Shumilina E, Lang F, Föller M. AMP-activated protein kinase regulates hERG potassium channel. Pflugers Arch 2013; 465:1573-82. [PMID: 23716168 DOI: 10.1007/s00424-013-1299-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 05/14/2013] [Indexed: 01/07/2023]
Abstract
Besides their role in cardiac repolarization, human ether-a-go-go-related gene potassium (hERG) channels are expressed in several tumor cells including rhabdomyosarcoma cells. The channels foster cell proliferation. Ubiquitously expressed AMP-dependent protein kinase (AMPK) is a serine-/threonine kinase, stimulating energy-generating and inhibiting energy-consuming processes thereby helping cells survive periods of energy depletion. AMPK has previously been shown to regulate Na⁺/K⁺ ATPase, Na⁺/Ca²⁺ exchangers, Ca²⁺ channels and K⁺ channels. The present study tested whether AMPK regulates hERG channel activity. Wild type AMPK (α1β1γ1), constitutively active (γR70Q)AMPK (α1β1γ1(R70Q)), or catalytically inactive (αK45R)AMPK (α1(K45R)β1γ1) were expressed in Xenopus oocytes with hERG. Tail currents were determined as a measure of hERG channel activity by two-electrode-voltage clamp. hERG membrane abundance was quantified by chemiluminescence and visualized by immunocytochemistry and confocal microscopy. Moreover, hERG currents were measured in RD rhabdomyosarcoma cells after pharmacological modification of AMPK activity using the patch clamp technique. Coexpression of wild-type AMPK and of constitutively active (γR70Q)AMPK significantly downregulated the tail currents in hERG-expressing Xenopus oocytes. Pharmacological activation of AMPK with AICAR or with phenformin inhibited hERG currents in Xenopus oocytes, an effect abrogated by AMPK inhibitor compound C. (γR70Q)AMPK enhanced the Nedd4-2-dependent downregulation of hERG currents. Coexpression of constitutively active (γR70Q)AMPK decreased membrane expression of hERG in Xenopus oocytes. Compound C significantly enhanced whereas AICAR tended to inhibit hERG currents in RD rhabdomyosarcoma cells. AMPK is a powerful regulator of hERG-mediated currents in both, Xenopus oocytes and RD rhabdomyosarcoma cells. AMPK-dependent regulation of hERG may be particularly relevant in cardiac hypertrophy and tumor growth.
Collapse
Affiliation(s)
- Ahmad Almilaji
- Department of Physiology, University of Tübingen, Gmelinstr. 5, 72076, Tübingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lamothe SM, Zhang S. The serum- and glucocorticoid-inducible kinases SGK1 and SGK3 regulate hERG channel expression via ubiquitin ligase Nedd4-2 and GTPase Rab11. J Biol Chem 2013; 288:15075-84. [PMID: 23589291 DOI: 10.1074/jbc.m113.453670] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The hERG (human ether-a-go-go-related gene) encodes the α subunit of the rapidly activating delayed rectifier potassium channel (IKr). Dysfunction of hERG channels due to mutations or certain medications causes long QT syndrome, which can lead to fatal ventricular arrhythmias or sudden death. Although the abundance of hERG in the plasma membrane is a key determinant of hERG functionality, the mechanisms underlying its regulation are not well understood. In the present study, we demonstrated that overexpression of the stress-responsive serum- and glucocorticoid-inducible kinase (SGK) isoforms SGK1 and SGK3 increased the current and expression level of the membrane-localized mature proteins of hERG channels stably expressed in HEK 293 (hERG-HEK) cells. Furthermore, the synthetic glucocorticoid, dexamethasone, increased the current and abundance of mature ERG proteins in both hERG-HEK cells and neonatal cardiac myocytes through the enhancement of SGK1 but not SGK3 expression. We have previously shown that mature hERG channels are degraded by ubiquitin ligase Nedd4-2 via enhanced channel ubiquitination. Here, we showed that SGK1 or SGK3 overexpression increased Nedd4-2 phosphorylation, which is known to inhibit Nedd4-2 activity. Nonetheless, disruption of the Nedd4-2 binding site in hERG channels did not eliminate the SGK-induced increase in hERG expression. Additional disruption of Rab11 proteins led to a complete elimination of SGK-mediated increase in hERG expression. These results show that SGK enhances the expression level of mature hERG channels by inhibiting Nedd4-2 as well as by promoting Rab11-mediated hERG recycling.
Collapse
Affiliation(s)
- Shawn M Lamothe
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | | |
Collapse
|
50
|
Abstract
KLHL3 is a BTB-BACK-Kelch family protein that serves as a substrate adapter in Cullin3 (Cul3) E3 ubiquitin ligase complexes. KLHL3 is highly expressed in distal nephron tubules where it is involved in the regulation of electrolyte homeostasis and blood pressure. Mutations in KLHL3 have been identified in patients with inherited hypertension disorders, and several of the disease-associated mutations are located in the presumed Cul3 binding region. Here, we report the crystal structure of a complex between the KLHL3 BTB-BACK domain dimer and two copies of an N terminal fragment of Cul3. We use isothermal titration calorimetry to directly demonstrate that several of the disease mutations in the KLHL3 BTB-BACK domains disrupt the association with Cul3. Both the BTB and BACK domains contribute to the Cul3 interaction surface, and an extended model of the dimeric CRL3 complex places the two E2 binding sites in a suprafacial arrangement with respect to the presumed substrate-binding sites.
Collapse
Affiliation(s)
- Alan X. Ji
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Gilbert G. Privé
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Ontario Cancer Institute, Campbell Family Institute for Cancer Research, University Health Network, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|