1
|
Yuan Y, Qian L, Miao Y, Cui Q, Cao T, Yu Y, Zhang T, Zhao Q, Zhang R, Ren T, Zuo Y, Du Q, Qiao C, Wu Q, Zheng Z, Li M, Chinn YE, Xu W, Peng T, Chen R, Xiong S, Zheng H. Targeting Viperin prevents coxsackievirus B3-induced acute heart failure. Cell Discov 2025; 11:34. [PMID: 40195316 PMCID: PMC11977219 DOI: 10.1038/s41421-025-00778-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/21/2025] [Indexed: 04/09/2025] Open
Abstract
Coxsackievirus B3 (CVB3)-induced acute heart failure (AHF) is a common cause of cardiogenic death in young- and middle-aged people. However, the key molecular events linking CVB3 to AHF remain largely unknown, resulting in a lack of targeted therapy strategies thus far. Here, we unexpectedly found that Viperin deficiency does not promote CVB3 infection but protects mice from CVB3-induced AHF. Importantly, cardiac-specific expression of Viperin can induce cardiac dysfunction. Mechanistically, CVB3-encoded 3C protease rescues Viperin protein expression in cardiomyocytes by lowering UBE4A. Viperin in turn interacts with and reduces STAT1 to activate SGK1-KCNQ1 signaling, and eventually leads to cardiac electrical dysfunction and subsequent AHF. Furthermore, we designed an interfering peptide VS-IP1, which blocked Viperin-mediated STAT1 degradation and therefore prevented CVB3-induced AHF. This study established the first signaling link between CVB3 and cardiac electrical dysfunction, and revealed the potential of interfering peptides targeting Viperin for the treatment of CVB3-induced AHF.
Collapse
Affiliation(s)
- Yukang Yuan
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Liping Qian
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Ying Miao
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Qun Cui
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Ting Cao
- Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Yong Yu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tingting Zhang
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Qian Zhao
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Renxia Zhang
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Tengfei Ren
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Yibo Zuo
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Qian Du
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Caixia Qiao
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Qiuyu Wu
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Zhijin Zheng
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Minqi Li
- Medical College of Nantong University, Nantong, Jiangsu, China
| | - Y Eugene Chinn
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Wei Xu
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Tianqing Peng
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada
- Department of Medicine, Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Ruizhen Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Sidong Xiong
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China.
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China.
| | - Hui Zheng
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China.
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Vanoye CG, Desai RR, John JD, Hoffman SC, Fink N, Zhang Y, Venkatesh OG, Roe J, Adusumilli S, Jairam NP, Sanders CR, Gordon AS, George AL. Functional profiling of KCNE1 variants informs population carrier frequency of Jervell and Lange-Nielsen syndrome type 2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.646046. [PMID: 40236191 PMCID: PMC11996308 DOI: 10.1101/2025.03.28.646046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Congenital long-QT syndrome (LQTS) is most often associated with pathogenic variants in KCNQ1 encoding the pore-forming voltage-gated potassium channel subunit of the slow delayed rectifier current ( I Ks ). Generation of I Ks requires assembly of KCNQ1 with an auxiliary subunit encoded by KCNE1 , which is also associated with LQTS but causality of autosomal dominant disease is disputed. By contrast, KCNE1 is an accepted cause of recessive type 2 Jervell and Lange-Nielson syndrome (JLN2). The functional consequences of most KCNE1 variants have not been determined and the population prevalence of JLN2 is unknown. Methods : We determined the functional properties of 95 KCNE1 variants co-expressed with KCNQ1 in heterologous cells using high-throughput voltage-clamp recording. Experiments were conducted with each KCNE1 variant expressed in the homozygous state and then a subset was studied in the heterozygous state. The carrier frequency of JLN2 was estimated by considering the population prevalence of dysfunctional variants. Results : There is substantial overlap between disease-associated and population KCNE1 variants. When examined in the homozygous state, 68 KCNE1 variants exhibited significant differences in at least one functional property compared to WT KCNE1, whereas 27 variants did not significantly affect function. Most dysfunctional variants exhibited loss-of-function properties. We observed no evidence of dominant-negative effects. Most variants were scored as variants of uncertain significance (VUS) and inclusion of functional data resulted in revised classifications for only 14 variants. The population carrier frequency of JLN2 was calculated as 1 in 1034. Peak current density and activation voltage-dependence but no other biophysical properties were correlated with findings from a mutational scan of KCNE1. Conclusions : Among 95 disease-associated or population KCNE1 variants, many exhibit abnormal functional properties but there was no evidence of dominant-negative behaviors. Using functional data, we inferred a population carrier frequency for recessive JLN2. This work helps clarify the pathogenicity of KCNE1 variants.
Collapse
|
3
|
Caudal A, Liu Y, Pang PD, Maison DP, Nakasuka K, Feng J, Schwarzer-Sperber HS, Schwarzer R, Moffatt E, Henrich TJ, Padmanabhan A, Connolly AJ, Wu JC, Tseng ZH. Transcriptomic Profiling of Human Myocardium at Sudden Death to Define Vulnerable Substrate for Lethal Arrhythmias. JACC Clin Electrophysiol 2025; 11:143-155. [PMID: 39545913 PMCID: PMC11809765 DOI: 10.1016/j.jacep.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND While some chronic pathological substrates for sudden cardiac death (SCD) are well known (eg, coronary artery disease and left ventricular [LV] dysfunction), the acute vulnerable myocardial state predisposing to fatal arrhythmia remains a critical barrier to near-term SCD prevention. OBJECTIVES This study sought to define the distinct myocardial transcriptomic profile of autopsy-defined arrhythmic sudden deaths, compared to nonarrhythmic sudden deaths and trauma deaths, to determine the acute vulnerable state in the hours to days before SCD. METHODS We used autopsy to adjudicate arrhythmic from nonarrhythmic causes in 1,265 sudden deaths in San Francisco County from 2011 to 2018. We performed a degradation-tolerant transcriptomic evaluation of LVs sampled at the time of SCD from 245 consented cases using a curated panel of 448 gene probes with known or hypothesized association with SCD. RESULTS The targeted transcriptome of arrhythmic (n = 129) vs nonarrhythmic (n = 90 nonarrhythmic sudden deaths + 26 trauma deaths) LV samples revealed 31 differentially up-regulated and 36 down-regulated genes (adjusted P < 0.05) related to the collagen-containing extracellular matrix (up-regulation of FAP, FMOD, and LTBP2), regulation of ion transport (up-regulation of KCNA5 and KCNN3 and down-regulation of KCNJ8, KCNK1, and KCNJ5), and contraction (down-regulation of MYH6). Fibrosis-related genes showed the highest magnitude increased expression in arrhythmic vs nonarrhythmic deaths and vs published transcriptomes from end-stage heart failure. After molecular stratification by known markers for mature (COL1A1, COL1A2, COL3A1) and active (POSTN, MEOX1) fibrosis, cases with the highest expression of both had the largest proportion of arrhythmic cause of death (n = 27 of 36 [75%]) vs cases with low expression of both markers (n = 87 of 181 [38%]) (P = 0.006) or vs mature only (n = 10 of 14 [71%]) or active only (n = 5 of 14 [36%]). Activated fibroblast gene expression signature was enriched in arrhythmic female vs arrhythmic male cases, among other sex-specific differences in ion-channel and myosin (up-regulation of SCN4B, SCN8A, and KCNAB1 in females and KCNJ4 and MYH7B in males) expression. CONCLUSIONS RNA profiling of the myocardium at SCD identifies active fibrosis, undetectable by conventional clinical methods, in the presence of fixed scar and selected ion-channel dysregulation (more pronounced among female cases) as an acute vulnerable substrate for fatal arrhythmias. These findings may represent novel directions to identify patients at elevated near-term risk for SCD and critical pathways for intervention to reduce acute lethal arrhythmias.
Collapse
Affiliation(s)
- Arianne Caudal
- Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Yu Liu
- Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Paul D Pang
- Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - David P Maison
- Division of Experimental Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Kosuke Nakasuka
- Cardiac Electrophysiology, Cardiology Division, School of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Jean Feng
- Department of Epidemiology and Biostatistics, School of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - H S Schwarzer-Sperber
- Institute for the Research on HIV and AIDS-Associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Roland Schwarzer
- Institute for the Research on HIV and AIDS-Associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ellen Moffatt
- Office of the Chief Medical Examiner, City and County of San Francisco, San Francisco, California, USA
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Arun Padmanabhan
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA; Gladstone Institute for Cardiovascular Disease, San Francisco, California, USA; Chan Zuckerberg Biohub San Francisco, California, USA
| | - Andrew J Connolly
- Department of Pathology, School of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Zian H Tseng
- Cardiac Electrophysiology, Cardiology Division, School of Medicine, University of California-San Francisco, San Francisco, California, USA; Department of Medicine, University of California-San Francisco, San Francisco, California, USA.
| |
Collapse
|
4
|
Giannetti F, Barbieri M, Shiti A, Casini S, Sager PT, Das S, Pradhananga S, Srinivasan D, Nimani S, Alerni N, Louradour J, Mura M, Gnecchi M, Brink P, Zehender M, Koren G, Zaza A, Crotti L, Wilde AAM, Schwartz PJ, Remme CA, Gepstein L, Sala L, Odening KE. Gene- and variant-specific efficacy of serum/glucocorticoid-regulated kinase 1 inhibition in long QT syndrome types 1 and 2. Europace 2023; 25:euad094. [PMID: 37099628 PMCID: PMC10228615 DOI: 10.1093/europace/euad094] [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: 02/18/2023] [Accepted: 03/20/2023] [Indexed: 04/28/2023] Open
Abstract
AIMS Current long QT syndrome (LQTS) therapy, largely based on beta-blockade, does not prevent arrhythmias in all patients; therefore, novel therapies are warranted. Pharmacological inhibition of the serum/glucocorticoid-regulated kinase 1 (SGK1-Inh) has been shown to shorten action potential duration (APD) in LQTS type 3. We aimed to investigate whether SGK1-Inh could similarly shorten APD in LQTS types 1 and 2. METHODS AND RESULTS Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and hiPSC-cardiac cell sheets (CCS) were obtained from LQT1 and LQT2 patients; CMs were isolated from transgenic LQT1, LQT2, and wild-type (WT) rabbits. Serum/glucocorticoid-regulated kinase 1 inhibition effects (300 nM-10 µM) on field potential durations (FPD) were investigated in hiPSC-CMs with multielectrode arrays; optical mapping was performed in LQT2 CCS. Whole-cell and perforated patch clamp recordings were performed in isolated LQT1, LQT2, and WT rabbit CMs to investigate SGK1-Inh (3 µM) effects on APD. In all LQT2 models across different species (hiPSC-CMs, hiPSC-CCS, and rabbit CMs) and independent of the disease-causing variant (KCNH2-p.A561V/p.A614V/p.G628S/IVS9-28A/G), SGK1-Inh dose-dependently shortened FPD/APD at 0.3-10 µM (by 20-32%/25-30%/44-45%). Importantly, in LQT2 rabbit CMs, 3 µM SGK1-Inh normalized APD to its WT value. A significant FPD shortening was observed in KCNQ1-p.R594Q hiPSC-CMs at 1/3/10 µM (by 19/26/35%) and in KCNQ1-p.A341V hiPSC-CMs at 10 µM (by 29%). No SGK1-Inh-induced FPD/APD shortening effect was observed in LQT1 KCNQ1-p.A341V hiPSC-CMs or KCNQ1-p.Y315S rabbit CMs at 0.3-3 µM. CONCLUSION A robust SGK1-Inh-induced APD shortening was observed across different LQT2 models, species, and genetic variants but less consistently in LQT1 models. This suggests a genotype- and variant-specific beneficial effect of this novel therapeutic approach in LQTS.
Collapse
Affiliation(s)
- Federica Giannetti
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Miriam Barbieri
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Assad Shiti
- Rappaport Faculty of Medicine and Research Institute, Technion–Israel Institute of Technology, Haifa, Israel
| | - Simona Casini
- Amsterdam UMC Location AMC Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam, The Netherlands
| | - Philip T Sager
- Thryv Therapeutics Inc., Montreal, Canada
- Cardiovascular Research Institute, Stanford University, Palo Alto, CA, USA
| | - Saumya Das
- Thryv Therapeutics Inc., Montreal, Canada
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Saranda Nimani
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Nicolò Alerni
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Julien Louradour
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Manuela Mura
- Department of Cardiothoracic and Vascular Sciences–Translational Cardiology Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Massimiliano Gnecchi
- Department of Cardiothoracic and Vascular Sciences–Translational Cardiology Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Pavia, Italy
| | - Paul Brink
- Department of Medicine, University of Stellenbosch, Tygerberg, South Africa
| | - Manfred Zehender
- Department of Cardiology and Angiology I, University Heart Center Freiburg, University Medical Center Freiburg, Freiburg, Germany
| | - Gideon Koren
- Cardiovascular Research Center, Brown University, Providence, RI, USA
| | - Antonio Zaza
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Lia Crotti
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Arthur A M Wilde
- Amsterdam UMC Location AMC Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam, The Netherlands
| | - Peter J Schwartz
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Carol Ann Remme
- Amsterdam UMC Location AMC Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam, The Netherlands
| | - Lior Gepstein
- Rappaport Faculty of Medicine and Research Institute, Technion–Israel Institute of Technology, Haifa, Israel
- Cardiology Department, Rambam Health Care Campus, Haifa, Israel
| | - Luca Sala
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Katja E Odening
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| |
Collapse
|
5
|
Dellin M, Rohrbeck I, Asrani P, Schreiber JA, Ritter N, Glorius F, Wünsch B, Budde T, Temme L, Strünker T, Stallmeyer B, Tüttelmann F, Meuth SG, Spehr M, Matschke J, Steinbicker A, Gatsogiannis C, Stoll R, Strutz-Seebohm N, Seebohm G. The second PI(3,5)P 2 binding site in the S0 helix of KCNQ1 stabilizes PIP 2-at the primary PI1 site with potential consequences on intermediate-to-open state transition. Biol Chem 2023; 404:241-254. [PMID: 36809224 DOI: 10.1515/hsz-2022-0247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/13/2022] [Indexed: 02/23/2023]
Abstract
The Phosphatidylinositol 3-phosphate 5-kinase Type III PIKfyve is the main source for selectively generated phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), a known regulator of membrane protein trafficking. PI(3,5)P2 facilitates the cardiac KCNQ1/KCNE1 channel plasma membrane abundance and therewith increases the macroscopic current amplitude. Functional-physical interaction of PI(3,5)P2 with membrane proteins and its structural impact is not sufficiently understood. This study aimed to identify molecular interaction sites and stimulatory mechanisms of the KCNQ1/KCNE1 channel via the PIKfyve-PI(3,5)P2 axis. Mutational scanning at the intracellular membrane leaflet and nuclear magnetic resonance (NMR) spectroscopy identified two PI(3,5)P2 binding sites, the known PIP2 site PS1 and the newly identified N-terminal α-helix S0 as relevant for functional PIKfyve effects. Cd2+ coordination to engineered cysteines and molecular modeling suggest that repositioning of S0 stabilizes the channel s open state, an effect strictly dependent on parallel binding of PI(3,5)P2 to both sites.
Collapse
Affiliation(s)
- Maurice Dellin
- IfGH-Cellular Electrophysiology, Department of Cardiology and Angiology, University Hospital of Münster, Robert-Koch Str. 45, D-48149, Münster, Germany
| | - Ina Rohrbeck
- IfGH-Cellular Electrophysiology, Department of Cardiology and Angiology, University Hospital of Münster, Robert-Koch Str. 45, D-48149, Münster, Germany
| | - Purva Asrani
- Faculty of Chemistry and Biochemistry, Biomolecular NMR Spectroscopy and RUBiospek|NMR, Ruhr University of Bochum, Universitätsstraße 150, D-44780, Bochum, Germany
| | - Julian A Schreiber
- IfGH-Cellular Electrophysiology, Department of Cardiology and Angiology, University Hospital of Münster, Robert-Koch Str. 45, D-48149, Münster, Germany
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Nadine Ritter
- IfGH-Cellular Electrophysiology, Department of Cardiology and Angiology, University Hospital of Münster, Robert-Koch Str. 45, D-48149, Münster, Germany
- GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Frank Glorius
- GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, Münster, Germany
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster, Corrensstrasse 40, D-48149, Münster, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
- GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Thomas Budde
- GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, Münster, Germany
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149, Münster, Germany
| | - Louisa Temme
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
- GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Timo Strünker
- GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, Münster, Germany
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Domagkstraße 11, D-48149, Münster, Germany
- Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany
| | - Birgit Stallmeyer
- Institute of Reproductive Genetics, University of Münster, Vesaliusweg 12-14, D-48149, Münster, Germany
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Vesaliusweg 12-14, D-48149, Münster, Germany
| | - Sven G Meuth
- Department of Neurology, Heinrich Heine University Düsseldorf, Moorenstraße 5, D-40225, Düsseldorf, Germany
| | - Marc Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Worringerweg 3, D-52074, Aachen, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, D-45147, Essen, Germany
| | - Andrea Steinbicker
- Goethe University Frankfurt and University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590, Frankfurt, Germany
| | - Christos Gatsogiannis
- Institute for Medical Physics and Biophysics and Center for Soft Nanoscience, Westfälische Wilhelms-Universität Münster, Münster, Busso-Peus Strasse 10, D-48149, Germany
| | - Raphael Stoll
- Faculty of Chemistry and Biochemistry, Biomolecular NMR Spectroscopy and RUBiospek|NMR, Ruhr University of Bochum, Universitätsstraße 150, D-44780, Bochum, Germany
| | - Nathalie Strutz-Seebohm
- IfGH-Cellular Electrophysiology, Department of Cardiology and Angiology, University Hospital of Münster, Robert-Koch Str. 45, D-48149, Münster, Germany
| | - Guiscard Seebohm
- IfGH-Cellular Electrophysiology, Department of Cardiology and Angiology, University Hospital of Münster, Robert-Koch Str. 45, D-48149, Münster, Germany
- GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, Münster, Germany
| |
Collapse
|
6
|
Nagata Y, Watanabe R, Eichhorn C, Ohno S, Aiba T, Ishikawa T, Nakano Y, Aizawa Y, Hayashi K, Murakoshi N, Nakajima T, Yagihara N, Mishima H, Sudo T, Higuchi C, Takahashi A, Sekine A, Makiyama T, Tanaka Y, Watanabe A, Tachibana M, Morita H, Yoshiura KI, Tsunoda T, Watanabe H, Kurabayashi M, Nogami A, Kihara Y, Horie M, Shimizu W, Makita N, Tanaka T. Targeted deep sequencing analyses of long QT syndrome in a Japanese population. PLoS One 2022; 17:e0277242. [PMID: 36480497 PMCID: PMC9731492 DOI: 10.1371/journal.pone.0277242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/22/2022] [Indexed: 12/13/2022] Open
Abstract
Long QT syndrome (LQTS) is one of the most common inherited arrhythmias and multiple genes have been reported as causative. Presently, genetic diagnosis for LQTS patients is becoming widespread and contributing to implementation of therapies. However, causative genetic mutations cannot be detected in about 20% of patients. To elucidate additional genetic mutations in LQTS, we performed deep-sequencing of previously reported 15 causative and 85 candidate genes for this disorder in 556 Japanese LQTS patients. We performed in-silico filtering of the sequencing data and found 48 novel variants in 33 genes of 53 cases. These variants were predicted to be damaging to coding proteins or to alter the binding affinity of several transcription factors. Notably, we found that most of the LQTS-related variants in the RYR2 gene were in the large cytoplasmic domain of the N-terminus side. They might be useful for screening of LQTS patients who had no known genetic factors. In addition, when the mechanisms of these variants in the development of LQTS are revealed, it will be useful for early diagnosis, risk stratification, and selection of treatment.
Collapse
Affiliation(s)
- Yuki Nagata
- Bioresourse Research Center, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Ryo Watanabe
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Christian Eichhorn
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Seiko Ohno
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Takeshi Aiba
- Devision of Arrhythmia, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Taisuke Ishikawa
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University, Hiroshima, Japan
| | - Yoshiyasu Aizawa
- Department of Cardiology, International University of Health and Welfare Narita Hospital, Narita, Japan
| | - Kenshi Hayashi
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Nobuyuki Murakoshi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tadashi Nakajima
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Nobue Yagihara
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroyuki Mishima
- Department of Human Genetics, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Takeaki Sudo
- Institute of Education, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Chihiro Higuchi
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Atsushi Takahashi
- Department of Genomic Medicine, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Akihiro Sekine
- Department of Infection and Host Defense, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takeru Makiyama
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihiro Tanaka
- Center for Arrhythmia Research, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Atsuyuki Watanabe
- Department of Cardiology, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Motomi Tachibana
- Department of Cardiology, Sakakibara heart institute of Okayama, Okayama, Japan
| | - Hiroshi Morita
- Department of Cardiovascular Therapeutics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koh-ichiro Yoshiura
- Department of Human Genetics, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
- Division of Advanced Preventive Medical Sciences and Leading Medical Research Core Unit, Nagasaki Univerisity Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tatsuhiko Tsunoda
- Laboratory for Medical Science Mathematics, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hiroshi Watanabe
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akihiko Nogami
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Hiroshima University, Hiroshima, Japan
| | - Minoru Horie
- Department of Cardiovascular Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - Naomasa Makita
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Toshihiro Tanaka
- Bioresourse Research Center, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
7
|
Abstract
Coronavirus disease 2019 (COVID-19) has encompassed the globe since it was first observed just under 2 years ago. Although the disease is predominantly a respiratory illness, there have been observed complications throughout the various organ systems. Namely, cardiovascular complications, and, more specifically, arrhythmic complications have been described throughout the pandemic in patients with COVID-19. Management of atrial arrhythmias, ventricular arrhythmias, and bradyarrhythmias in patients with COVID-19 infection has been largely guided by our prior experience in the management of these arrhythmias in similar patient populations without infection. However, this review aims to highlight the specific considerations as they pertain to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the various arrhythmic manifestations observed with this disease.
Collapse
Affiliation(s)
| | | | - Elaine Y. Wan
- Address reprint requests and correspondence: Dr Elaine Wan, MD, FACC, FAHA, FHRS, Esther Aboodi Associate Professor of Medicine, 622 W 168th St, PH 3-Center, New York, NY 10032.
| |
Collapse
|
8
|
Andrzejewska M, Żebrowski JJ, Rams K, Ozimek M, Baranowski R. Assessment of time irreversibility in a time series using visibility graphs. FRONTIERS IN NETWORK PHYSIOLOGY 2022; 2:877474. [PMID: 36926071 PMCID: PMC10013024 DOI: 10.3389/fnetp.2022.877474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/24/2022] [Indexed: 11/07/2022]
Abstract
In this paper, we studied the time-domain irreversibility of time series, which is a fundamental property of systems in a nonequilibrium state. We analyzed a subgroup of the databases provided by University of Rochester, namely from the THEW Project. Our data consists of LQTS (Long QT Syndrome) patients and healthy persons. LQTS may be associated with an increased risk of sudden cardiac death (SCD), which is still a big clinical problem. ECG-based artificial intelligence methods can identify sudden cardiac death with a high accuracy. It follows that heart rate variability contains information about the possibility of SCD, which may be extracted, provided that appropriate methods are developed for this purpose. Our aim was to assess the complexity of both groups using visibility graph (VG) methods. Multivariate analysis of connection patterns of graphs built from time series was performed using multiplex visibility graph methods. For univariate time series, time irreversibility of the ECG interval QT of patients with LQTS was lower than for the healthy. However, we did not observe statistically significant difference in the comparison of RR intervals time series of the two groups studied. The connection patterns retrieved from multiplex VGs have more similarity with each other in the case of LQTS patients. This observation may be used to develop better methods for SCD risk stratification.
Collapse
Affiliation(s)
- Małgorzata Andrzejewska
- Cardiovascular Physics Group, Physics of Complex Systems Division, Faculty of Physics, Warsaw University of Technology, Warszawa, Poland
| | - Jan J Żebrowski
- Cardiovascular Physics Group, Physics of Complex Systems Division, Faculty of Physics, Warsaw University of Technology, Warszawa, Poland
| | - Karolina Rams
- Cardiovascular Physics Group, Physics of Complex Systems Division, Faculty of Physics, Warsaw University of Technology, Warszawa, Poland
| | - Mateusz Ozimek
- Cardiovascular Physics Group, Physics of Complex Systems Division, Faculty of Physics, Warsaw University of Technology, Warszawa, Poland
| | | |
Collapse
|
9
|
Noor S, Mohammad T, Ashraf GM, Farhat J, Bilgrami AL, Eapen MS, Sohal SS, Yadav DK, Hassan MI. Mechanistic insights into the role of serum-glucocorticoid kinase 1 in diabetic nephropathy: A systematic review. Int J Biol Macromol 2021; 193:562-573. [PMID: 34715204 DOI: 10.1016/j.ijbiomac.2021.10.165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/13/2022]
Abstract
Aberrant expression of serum-glucocorticoid kinase 1 (SGK1) contributes to the pathogenesis of multiple disorders, including diabetes, hypertension, obesity, fibrosis, and metabolic syndrome. SGK1 variant is expressed in the presence of insulin and several growth factors, eventually modulating various ion channels, carrier proteins, and transcription factors. SGK1 also regulates the enzymatic activity of Na+ K+ ATPase, glycogen synthase kinase-3, ubiquitin ligase Nedd4-2, and phosphomannose mutase impacting cell cycle regulation, neuroexcitation, and apoptosis. Ample evidence supports the crucial role of aberrant SGK1 expression in hyperglycemia-mediated secondary organ damage. Diabetic nephropathy (DN), a dreadful microvascular complication of diabetes, is the leading cause of end-stage renal failures with high morbidity and mortality rate. The complex pathogenesis of DN encompasses several influencing factors, including transcriptional factors, inflammatory markers, cytokines, epigenetic modulators, and abnormal enzymatic activities. SGK1 plays a pivotal role by controlling various physiological functions associated with the occurrence and progression of DN; therefore, targeting SGK1 may favorably influence the clinical outcome in patients with DN. This review aimed to provide mechanistic insights into SGK1 regulated DN pathogenesis and summarize the evidence supporting the therapeutic potential of SGK1 inhibition and its consequences on human health.
Collapse
Affiliation(s)
- Saba Noor
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam M Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Joviana Farhat
- College of Pharmacy, Al Ain University, Abu Dhabi 112612, United Arab Emirates
| | - Anwar L Bilgrami
- Deanship of Scientific Research, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Entomology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Mathew Suji Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Dharmendra Kumar Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, Yeonsu-gu, Incheon City 21924, South Korea.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
10
|
Martin-Batista E, Manville RW, Rivero-Pérez B, Bartolomé-Martín D, Alvarez de la Rosa D, Abbott GW, Giraldez T. Activation of SGK1.1 Upregulates the M-current in the Presence of Epilepsy Mutations. Front Mol Neurosci 2021; 14:798261. [PMID: 34899186 PMCID: PMC8662703 DOI: 10.3389/fnmol.2021.798261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
In the central nervous system, the M-current plays a critical role in regulating subthreshold electrical excitability of neurons, determining their firing properties and responsiveness to synaptic input. The M-channel is mainly formed by subunits Kv7.2 and Kv7.3 that co-assemble to form a heterotetrametric channel. Mutations in Kv7.2 and Kv7.3 are associated with hyperexcitability phenotypes including benign familial neonatal epilepsy (BFNE) and neonatal epileptic encephalopathy (NEE). SGK1.1, the neuronal isoform of the serum and glucocorticoids-regulated kinase 1 (SGK1), increases M-current density in neurons, leading to reduced excitability and protection against seizures. Herein, using two-electrode voltage clamp on Xenopus laevis oocytes, we demonstrate that SGK1.1 selectively activates heteromeric Kv7 subunit combinations underlying the M-current. Importantly, activated SGK1.1 increases M-channel activity in the presence of two different epilepsy mutations found in Kv7.2, R207W and A306T. In addition, proximity ligation assays in the N2a cell line allowed us to address the effect of these mutations on Kv7-SGK1.1-Nedd4 molecular associations, a proposed pathway underlying augmentation of M-channel activity by SGK1.1.
Collapse
Affiliation(s)
- Elva Martin-Batista
- Departamento de Ciencias Medicas Basicas and Instituto de Tecnologias Biomedicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Belinda Rivero-Pérez
- Departamento de Ciencias Medicas Basicas and Instituto de Tecnologias Biomedicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - David Bartolomé-Martín
- Departamento de Ciencias Medicas Basicas and Instituto de Tecnologias Biomedicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Diego Alvarez de la Rosa
- Departamento de Ciencias Medicas Basicas and Instituto de Tecnologias Biomedicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Teresa Giraldez
- Departamento de Ciencias Medicas Basicas and Instituto de Tecnologias Biomedicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| |
Collapse
|
11
|
Abstract
The physiological heart function is controlled by a well-orchestrated interplay of different ion channels conducting Na+, Ca2+ and K+. Cardiac K+ channels are key players of cardiac repolarization counteracting depolarizating Na+ and Ca2+ currents. In contrast to Na+ and Ca2+, K+ is conducted by many different channels that differ in activation/deactivation kinetics as well as in their contribution to different phases of the action potential. Together with modulatory subunits these K+ channel α-subunits provide a wide range of repolarizing currents with specific characteristics. Moreover, due to expression differences, K+ channels strongly influence the time course of the action potentials in different heart regions. On the other hand, the variety of different K+ channels increase the number of possible disease-causing mutations. Up to now, a plethora of gain- as well as loss-of-function mutations in K+ channel forming or modulating proteins are known that cause severe congenital cardiac diseases like the long-QT-syndrome, the short-QT-syndrome, the Brugada syndrome and/or different types of atrial tachyarrhythmias. In this chapter we provide a comprehensive overview of different K+ channels in cardiac physiology and pathophysiology.
Collapse
|
12
|
Etheridge SP, Asaki SY. COVID-19 Infection and Corrected QT Interval Prolongation-Collateral Damage From Our Newest Enemy. JAMA Netw Open 2021; 4:e217192. [PMID: 33890995 DOI: 10.1001/jamanetworkopen.2021.7192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
13
|
Abstract
Kv7.1-Kv7.5 (KCNQ1-5) K+ channels are voltage-gated K+ channels with major roles in neurons, muscle cells and epithelia where they underlie physiologically important K+ currents, such as neuronal M current and cardiac IKs. Specific biophysical properties of Kv7 channels make them particularly well placed to control the activity of excitable cells. Indeed, these channels often work as 'excitability breaks' and are targeted by various hormones and modulators to regulate cellular activity outputs. Genetic deficiencies in all five KCNQ genes result in human excitability disorders, including epilepsy, arrhythmias, deafness and some others. Not surprisingly, this channel family attracts considerable attention as potential drug targets. Here we will review biophysical properties and tissue expression profile of Kv7 channels, discuss recent advances in the understanding of their structure as well as their role in various neurological, cardiovascular and other diseases and pathologies. We will also consider a scope for therapeutic targeting of Kv7 channels for treatment of the above health conditions.
Collapse
|
14
|
Xu Parks X, Qudsi H, Braun C, Lopes CMB. The auxiliary subunit KCNE1 regulates KCNQ1 channel response to sustained calcium-dependent PKC activation. PLoS One 2020; 15:e0237591. [PMID: 32833978 PMCID: PMC7446858 DOI: 10.1371/journal.pone.0237591] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/29/2020] [Indexed: 11/18/2022] Open
Abstract
The slow cardiac delayed rectifier current (IKs) is formed by KCNQ1 and KCNE1 subunits and is one of the major repolarizing currents in the heart. Decrease of IKs currents either due to inherited mutations or pathological remodeling is associated with increased risk for cardiac arrhythmias and sudden death. Ca2+-dependent PKC isoforms (cPKC) are chronically activated in heart disease and diabetes. Recently, we found that sustained stimulation of the calcium-dependent PKCβII isoform leads to decrease in KCNQ1 subunit membrane localization and KCNQ1/KCNE1 channel activity, although the role of KCNE1 in this regulation was not explored. Here, we show that the auxiliary KCNE1 subunit expression is necessary for channel internalization. A mutation in a KCNE1 phosphorylation site (KCNE1(S102A)) abolished channel internalization in both heterologous expression systems and cardiomyocytes. Altogether, our results suggest that KCNE1(S102) phosphorylation by PKCβII leads to KCNQ1/KCNE1 channel internalization in response to sustained PKC stimulus, while leaving KCNQ1 homomeric channels in the membrane. This preferential internalization is expected to have strong impact on cardiac repolarization. Our results suggest that KCNE1(S102) is an important anti-arrhythmic drug target to prevent IKs pathological remodeling leading to cardiac arrhythmias.
Collapse
Affiliation(s)
- Xiaorong Xu Parks
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - Haani Qudsi
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - Chen Braun
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - Coeli M. B. Lopes
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| |
Collapse
|
15
|
Estadella I, Pedrós-Gámez O, Colomer-Molera M, Bosch M, Sorkin A, Felipe A. Endocytosis: A Turnover Mechanism Controlling Ion Channel Function. Cells 2020; 9:E1833. [PMID: 32759790 PMCID: PMC7463639 DOI: 10.3390/cells9081833] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 01/08/2023] Open
Abstract
Ion channels (IChs) are transmembrane proteins that selectively drive ions across membranes. The function of IChs partially relies on their abundance and proper location in the cell, fine-tuned by the delicate balance between secretory, endocytic, and degradative pathways. The disruption of this balance is associated with several diseases, such as Liddle's and long QT syndromes. Because of the vital role of these proteins in human health and disease, knowledge of ICh turnover is essential. Clathrin-dependent and -independent mechanisms have been the primary mechanisms identified with ICh endocytosis and degradation. Several molecular determinants recognized by the cellular internalization machinery have been discovered. Moreover, specific conditions can trigger the endocytosis of many IChs, such as the activation of certain receptors, hypokalemia, and some drugs. Ligand-dependent receptor activation primarily results in the posttranslational modification of IChs and the recruitment of important mediators, such as β-arrestins and ubiquitin ligases. However, endocytosis is not a final fate. Once internalized into endosomes, IChs are either sorted to lysosomes for degradation or recycled back to the plasma membrane. Rab proteins are crucial participants during these turnover steps. In this review, we describe the major ICh endocytic pathways, the signaling inputs triggering ICh internalization, and the key mediators of this essential cellular process.
Collapse
Affiliation(s)
- Irene Estadella
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (I.E.); (O.P.-G.); (M.C.-M.); (M.B.)
| | - Oriol Pedrós-Gámez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (I.E.); (O.P.-G.); (M.C.-M.); (M.B.)
| | - Magalí Colomer-Molera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (I.E.); (O.P.-G.); (M.C.-M.); (M.B.)
| | - Manel Bosch
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (I.E.); (O.P.-G.); (M.C.-M.); (M.B.)
- Centres Científics i Tecnològics de la Universitat de Barcelona (CCiTUB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Alexander Sorkin
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (I.E.); (O.P.-G.); (M.C.-M.); (M.B.)
| |
Collapse
|
16
|
Ion Channels as Therapeutic Targets for Viral Infections: Further Discoveries and Future Perspectives. Viruses 2020; 12:v12080844. [PMID: 32756358 PMCID: PMC7472218 DOI: 10.3390/v12080844] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022] Open
Abstract
Ion channels play key roles in almost all facets of cellular physiology and have emerged as key host cell factors for a multitude of viral infections. A catalogue of ion channel-blocking drugs have been shown to possess antiviral activity, some of which are in widespread human usage for ion channel-related diseases, highlighting new potential for drug repurposing. The emergence of ion channel–virus interactions has also revealed the intriguing possibility that channelopathies may explain some commonly observed virus induced pathologies. This field is rapidly evolving and an up-to-date summary of new discoveries can inform future perspectives. We herein discuss the role of ion channels during viral lifecycles, describe the recently identified ion channel drugs that can inhibit viral infections, and highlight the potential contribution of ion channels to virus-mediated disease.
Collapse
|
17
|
Oliveras A, Serrano-Novillo C, Moreno C, de la Cruz A, Valenzuela C, Soeller C, Comes N, Felipe A. The unconventional biogenesis of Kv7.1-KCNE1 complexes. SCIENCE ADVANCES 2020; 6:eaay4472. [PMID: 32270035 PMCID: PMC7112945 DOI: 10.1126/sciadv.aay4472] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 01/09/2020] [Indexed: 06/11/2023]
Abstract
The potassium channel Kv7.1 associates with the KCNE1 regulatory subunit to trigger cardiac I Ks currents. Although the Kv7.1/KCNE1 complex has received much attention, the subcellular compartment hosting the assembly is the subject of ongoing debate. Evidence suggests that the complex forms either earlier in the endoplasmic reticulum or directly at the plasma membrane. Kv7.1 and KCNE1 mutations, responsible for long QT syndromes, impair association and traffic, thereby altering I Ks currents. We found that Kv7.1 and KCNE1 do not assemble in the first stages of their biogenesis. Data support an unconventional secretory pathway for Kv7.1-KCNE1 that bypasses Golgi. This route targets channels to endoplasmic reticulum-plasma membrane junctions, where Kv7.1-KCNE1 assemble. This mechanism helps to resolve the ongoing controversy about the subcellular compartment hosting the association. Our results also provide new insights into I Ks channel localization at endoplasmic reticulum-plasma membrane junctions, highlighting an alternative anterograde trafficking mechanism for oligomeric ion channels.
Collapse
Affiliation(s)
- Anna Oliveras
- Molecular Physiology Laboratory, Departamento de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Clara Serrano-Novillo
- Molecular Physiology Laboratory, Departamento de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Cristina Moreno
- National Institute of Neurological Disorders and Stroke (NIH), Bethesda, MD, USA
| | - Alicia de la Cruz
- Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Spanish Network for Biomedical Research in Cardiovascular Research (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Christian Soeller
- Living Systems Institute and Biomedical Physics, University of Exeter, Exeter, UK
| | - Núria Comes
- Departamento De Biomedicina, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departamento de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
18
|
Lee YK, Sala L, Mura M, Rocchetti M, Pedrazzini M, Ran X, Mak TSH, Crotti L, Sham PC, Torre E, Zaza A, Schwartz PJ, Tse HF, Gnecchi M. MTMR4 SNVs modulate ion channel degradation and clinical severity in congenital long QT syndrome: insights in the mechanism of action of protective modifier genes. Cardiovasc Res 2020; 117:767-779. [PMID: 32173736 PMCID: PMC7898949 DOI: 10.1093/cvr/cvaa019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/23/2019] [Accepted: 01/22/2020] [Indexed: 01/26/2023] Open
Abstract
Aims In long QT syndrome (LQTS) patients, modifier genes modulate the arrhythmic risk associated with a disease-causing mutation. Their recognition can improve risk stratification and clinical management, but their discovery represents a challenge. We tested whether a cellular-driven approach could help to identify new modifier genes and especially their mechanism of action. Methods and results We generated human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) from two patients carrying the same KCNQ1-Y111C mutation, but presenting opposite clinical phenotypes. We showed that the phenotype of the iPSC-CMs derived from the symptomatic patient is due to impaired trafficking and increased degradation of the mutant KCNQ1 and wild-type human ether-a-go-go-related gene. In the iPSC-CMs of the asymptomatic (AS) patient, the activity of an E3 ubiquitin-protein ligase (Nedd4L) involved in channel protein degradation was reduced and resulted in a decreased arrhythmogenic substrate. Two single-nucleotide variants (SNVs) on the Myotubularin-related protein 4 (MTMR4) gene, an interactor of Nedd4L, were identified by whole-exome sequencing as potential contributors to decreased Nedd4L activity. Correction of these SNVs by CRISPR/Cas9 unmasked the LQTS phenotype in AS cells. Importantly, the same MTMR4 variants were present in 77% of AS Y111C mutation carriers of a separate cohort. Thus, genetically mediated interference with Nedd4L activation seems associated with protective effects. Conclusion Our finding represents the first demonstration of the cellular mechanism of action of a protective modifier gene in LQTS. It provides new clues for advanced risk stratification and paves the way for the design of new therapies targeting this specific molecular pathway.
Collapse
Affiliation(s)
- Yee-Ki Lee
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Luca Sala
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milano, Italy.,Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Manuela Mura
- Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Cardiothoracic and Vascular Sciences, Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marcella Rocchetti
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Matteo Pedrazzini
- Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Xinru Ran
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong, Hong Kong SAR, China.,Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Timothy S H Mak
- Department of Psychiatry, The University of Hong Kong, Hong Kong SAR, China
| | - Lia Crotti
- Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy.,Department of Cardiovascular, Neural and Metabolic Sciences, Istituto Auxologico Italiano, IRCCS, San Luca Hospital, Milan, Italy.,Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Pak C Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong SAR, China.,Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.,State Key Laboratory for Cognitive and Brain Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Eleonora Torre
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Antonio Zaza
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Peter J Schwartz
- Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong, Hong Kong SAR, China.,Guangzhou Institutes of Biomedicine and Health, Guangzhou, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Hong Kong SAR, China
| | - Massimiliano Gnecchi
- Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Cardiothoracic and Vascular Sciences, Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Pavia, Italy.,Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
19
|
Ronzier E, Parks XX, Qudsi H, Lopes CM. Statin-specific inhibition of Rab-GTPase regulates cPKC-mediated IKs internalization. Sci Rep 2019; 9:17747. [PMID: 31780674 PMCID: PMC6882895 DOI: 10.1038/s41598-019-53700-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 10/21/2019] [Indexed: 12/18/2022] Open
Abstract
Statins are prescribed for prevention and treatment of coronary artery disease. Statins have different cholesterol lowering abilities, with rosuvastatin and atorvastatin being the most effective, while statins like simvastatin and fluvastatin having lower effectiveness. Statins, in addition to their cholesterol lowering effects, can prevent isoprenylation of Rab-GTPase proteins, a protein family important for the regulation of membrane-bound protein trafficking. Here we show that endosomal localization of Rab-GTPases (Rab5, Rab7 and Rab11) was inhibited in a statin-specific manner, with stronger effects by fluvastatin, followed by simvastatin and atorvastatin, and with a limited effect by rosuvastatin. Fluvastatin inhibition of Rab5 has been shown to mediate cPKC-dependent trafficking regulation of the cardiac delayed rectifier KCNQ1/KCNE1 channels. We observed statin-specific inhibition of channel regulation consistent with statin-specific Rab-GTPase inhibition both in heterologous systems and cardiomyocytes. Our results uncover a non-cholesterol-reducing statin-specific effect of statins. Because Rab-GTPases are important regulators of membrane trafficking they may underlie statin specific pleiotropic effects. Therefore, statin-specificity may allow better treatment tailoring.
Collapse
Affiliation(s)
- Elsa Ronzier
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Xiaorong Xu Parks
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Haani Qudsi
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Coeli M Lopes
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
20
|
Kurakami K, Norota I, Nasu F, Ohshima S, Nagasawa Y, Konno Y, Obara Y, Ishii K. KCNQ1 is internalized by activation of α1 adrenergic receptors. Biochem Pharmacol 2019; 169:113628. [DOI: 10.1016/j.bcp.2019.113628] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/30/2019] [Indexed: 01/25/2023]
|
21
|
Parks XX, Ronzier E, O-Uchi J, Lopes CM. Fluvastatin inhibits Rab5-mediated IKs internalization caused by chronic Ca 2+-dependent PKC activation. J Mol Cell Cardiol 2019; 129:314-325. [PMID: 30898664 DOI: 10.1016/j.yjmcc.2019.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 02/26/2019] [Accepted: 03/16/2019] [Indexed: 10/27/2022]
Abstract
Statins, in addition to their cholesterol lowering effects, can prevent isoprenylation of Rab GTPase proteins, a key protein family for the regulation of protein trafficking. Rab-GTPases have been shown to be involved in the control of membrane expression level of ion channels, including one of the major cardiac repolarizing channels, IKs. Decreased IKs function has been observed in a number of disease states and associated with increased propensity for arrhythmias, but the mechanism underlying IKs decrease remains elusive. Ca2+-dependent PKC isoforms (cPKC) are chronically activated in variety of human diseases and have been suggested to acutely regulate IKs function. We hypothesize that chronic cPKC stimulation leads to Rab-mediated decrease in IKs membrane expression, and that can be prevented by statins. In this study we show that chronic cPKC stimulation caused a dramatic Rab5 GTPase-dependent decrease in plasma membrane localization of the IKs pore forming subunit KCNQ1, reducing IKs function. Our data indicates fluvastatin inhibition of Rab5 restores channel localization and function after cPKC-mediated channel internalization. Our results indicate a novel statin anti-arrhythmic effect that would be expected to inhibit pathological electrical remodeling in a number of disease states associated with high cPKC activation. Because Rab-GTPases are important regulators of membrane trafficking they may underlie other statin pleiotropic effects.
Collapse
Affiliation(s)
- Xiaorong Xu Parks
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Elsa Ronzier
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Jin O-Uchi
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America; Lillehei Heart Institute, University of Minnesota, 2231 6th Street SE, Minneapolis, MN 55455, United States of America
| | - Coeli M Lopes
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America.
| |
Collapse
|
22
|
Locke MN, Thorner J. Rab5 GTPases are required for optimal TORC2 function. J Cell Biol 2019; 218:961-976. [PMID: 30578283 PMCID: PMC6400565 DOI: 10.1083/jcb.201807154] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 11/16/2018] [Accepted: 12/11/2018] [Indexed: 12/19/2022] Open
Abstract
Target of rapamycin complex-2 (TORC2), a conserved protein kinase complex, is an indispensable regulator of plasma membrane homeostasis. In budding yeast (Saccharomyces cerevisiae), the essential downstream effector of TORC2 is protein kinase Ypk1 and its paralog Ypk2. Muk1, a Rab5-specific guanine nucleotide exchange factor (GEF), was identified in our prior global screen for candidate Ypk1 targets. We confirm here that Muk1 is a substrate of Ypk1 and demonstrate that Ypk1-mediated phosphorylation stimulates Muk1 function in vivo. Strikingly, yeast lacking its two Rab5 GEFs (Muk1 and Vps9) or its three Rab5 paralogs (Vps21/Ypt51, Ypt52, and Ypt53) or overexpressing Msb3, a Rab5-directed GTPase-activating protein, all exhibit pronounced reduction in TORC2-mediated phosphorylation and activation of Ypk1. Vps21 coimmunoprecipitates with TORC2, and immuno-enriched TORC2 is less active in vitro in the absence of Rab5 GTPases. Thus, TORC2-dependent and Ypk1-mediated activation of Muk1 provides a control circuit for positive (self-reinforcing) up-regulation to sustain TORC2-Ypk1 signaling.
Collapse
Affiliation(s)
- Melissa N Locke
- Division of Biochemistry, Biophysics, and Structural Biology and Division of Cell and Developmental Biology, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| | - Jeremy Thorner
- Division of Biochemistry, Biophysics, and Structural Biology and Division of Cell and Developmental Biology, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
23
|
Barrese V, Stott JB, Figueiredo HB, Aubdool AA, Hobbs AJ, Jepps TA, McNeish AJ, Greenwood IA. Angiotensin II Promotes K V7.4 Channels Degradation Through Reduced Interaction With HSP90 (Heat Shock Protein 90). Hypertension 2018; 71:1091-1100. [PMID: 29686000 DOI: 10.1161/hypertensionaha.118.11116] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/16/2018] [Accepted: 03/22/2018] [Indexed: 12/11/2022]
Abstract
Voltage-gated Kv7.4 channels have been implicated in vascular smooth muscle cells' activity because they modulate basal arterial contractility, mediate responses to endogenous vasorelaxants, and are downregulated in several arterial beds in different models of hypertension. Angiotensin II (Ang II) is a key player in hypertension that affects the expression of several classes of ion channels. In this study, we evaluated the effects of Ang II on the expression and function of vascular Kv7.4. Western blot and quantitative polymerase chain reaction revealed that in whole rat mesenteric artery, Ang II incubation for 1 to 7 hours decreased Kv7.4 protein expression without reducing transcript levels. Moreover, Ang II decreased XE991 (Kv7)-sensitive currents and attenuated membrane potential hyperpolarization and relaxation induced by the Kv7 activator ML213. Ang II also reduced Kv7.4 staining at the plasma membrane of vascular smooth muscle cells. Proteasome inhibition with MG132 prevented Ang II-induced decrease of Kv7.4 levels and counteracted the functional impairment of ML213-induced relaxation in myography experiments. Proximity ligation assays showed that Ang II impaired the interaction of Kv7.4 with the molecular chaperone HSP90 (heat shock protein 90), enhanced the interaction of Kv7.4 with the E3 ubiquitin ligase CHIP (C terminus of Hsp70-interacting protein), and increased Kv7.4 ubiquitination. Similar alterations were found in mesenteric vascular smooth muscle cells isolated from Ang II-infused mice. The effect of Ang II was emulated by 17-AAG (17-demethoxy-17-(2-propenylamino) geldanamycin) that inhibits HSP90 interactions with client proteins. These results show that Ang II downregulates Kv7.4 by altering protein stability through a decrease of its interaction with HSP90. This leads to the recruitment of CHIP and Kv7.4 ubiquitination and degradation via the proteasome.
Collapse
Affiliation(s)
- Vincenzo Barrese
- From the Vascular Research Centre, Institute of Molecular and Clinical Sciences, St George's, University of London, United Kingdom (V.B., J.B.S., H.B.F., I.A.G.)
| | - Jennifer B Stott
- From the Vascular Research Centre, Institute of Molecular and Clinical Sciences, St George's, University of London, United Kingdom (V.B., J.B.S., H.B.F., I.A.G.)
| | - Hericka B Figueiredo
- From the Vascular Research Centre, Institute of Molecular and Clinical Sciences, St George's, University of London, United Kingdom (V.B., J.B.S., H.B.F., I.A.G.)
| | - Aisah A Aubdool
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary, University of London, United Kingdom (A.A.A., A.J.H.)
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary, University of London, United Kingdom (A.A.A., A.J.H.)
| | - Thomas A Jepps
- Department of Biomedical Sciences, University of Copenhagen, Denmark (T.A.J.)
| | - Alister J McNeish
- and Reading School of Pharmacy, University of Reading, United Kingdom (A.J.M.)
| | - Iain A Greenwood
- From the Vascular Research Centre, Institute of Molecular and Clinical Sciences, St George's, University of London, United Kingdom (V.B., J.B.S., H.B.F., I.A.G.)
| |
Collapse
|
24
|
Huang H, Kuenze G, Smith JA, Taylor KC, Duran AM, Hadziselimovic A, Meiler J, Vanoye CG, George AL, Sanders CR. Mechanisms of KCNQ1 channel dysfunction in long QT syndrome involving voltage sensor domain mutations. SCIENCE ADVANCES 2018; 4:eaar2631. [PMID: 29532034 PMCID: PMC5842040 DOI: 10.1126/sciadv.aar2631] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/02/2018] [Indexed: 05/21/2023]
Abstract
Mutations that induce loss of function (LOF) or dysfunction of the human KCNQ1 channel are responsible for susceptibility to a life-threatening heart rhythm disorder, the congenital long QT syndrome (LQTS). Hundreds of KCNQ1 mutations have been identified, but the molecular mechanisms responsible for impaired function are poorly understood. We investigated the impact of 51 KCNQ1 variants with mutations located within the voltage sensor domain (VSD), with an emphasis on elucidating effects on cell surface expression, protein folding, and structure. For each variant, the efficiency of trafficking to the plasma membrane, the impact of proteasome inhibition, and protein stability were assayed. The results of these experiments combined with channel functional data provided the basis for classifying each mutation into one of six mechanistic categories, highlighting heterogeneity in the mechanisms resulting in channel dysfunction or LOF. More than half of the KCNQ1 LOF mutations examined were seen to destabilize the structure of the VSD, generally accompanied by mistrafficking and degradation by the proteasome, an observation that underscores the growing appreciation that mutation-induced destabilization of membrane proteins may be a common human disease mechanism. Finally, we observed that five of the folding-defective LQTS mutant sites are located in the VSD S0 helix, where they interact with a number of other LOF mutation sites in other segments of the VSD. These observations reveal a critical role for the S0 helix as a central scaffold to help organize and stabilize the KCNQ1 VSD and, most likely, the corresponding domain of many other ion channels.
Collapse
Affiliation(s)
- Hui Huang
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37240, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Georg Kuenze
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240, USA
| | - Jarrod A. Smith
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37240, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Keenan C. Taylor
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37240, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Amanda M. Duran
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240, USA
| | - Arina Hadziselimovic
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37240, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240, USA
- Department of Bioinformatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Carlos G. Vanoye
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Alfred L. George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Charles R. Sanders
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37240, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
25
|
Barrese V, Stott JB, Greenwood IA. KCNQ-Encoded Potassium Channels as Therapeutic Targets. Annu Rev Pharmacol Toxicol 2018; 58:625-648. [DOI: 10.1146/annurev-pharmtox-010617-052912] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | | | - Iain A. Greenwood
- Vascular Biology Research Centre, Molecular and Clinical Sciences Institute, St George's, University of London, London, SW17 0RE, United Kingdom;, ,
| |
Collapse
|
26
|
Piccini I, Fehrmann E, Frank S, Müller FU, Greber B, Seebohm G. Adrenergic Stress Protection of Human iPS Cell-Derived Cardiomyocytes by Fast K v7.1 Recycling. Front Physiol 2017; 8:705. [PMID: 28959214 PMCID: PMC5603700 DOI: 10.3389/fphys.2017.00705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/31/2017] [Indexed: 01/09/2023] Open
Abstract
The fight-or-flight response (FFR), a physiological acute stress reaction, involves positive chronotropic and inotropic effects on heart muscle cells mediated through β-adrenoceptor activation. Increased systolic calcium is required to enable stronger heart contractions whereas elevated potassium currents are to limit the duration of the action potentials and prevent arrhythmia. The latter effect is accomplished by an increased functional activity of the Kv7.1 channel encoded by KCNQ1. Current knowledge, however, does not sufficiently explain the full extent of rapid Kv7.1 activation and may hence be incomplete. Using inducible genetic KCNQ1 complementation in KCNQ1-deficient human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), we here reinvestigate the functional role of Kv7.1 in adapting human CMs to adrenergic stress. Under baseline conditions, Kv7.1 was barely detectable at the plasma membrane of hiPSC-CMs, yet it fully protected these from adrenergic stress-induced beat-to-beat variability of repolarization and torsade des pointes-like arrhythmia. Furthermore, isoprenaline treatment increased field potential durations specifically in KCNQ1-deficient CMs to cause these adverse macroscopic effects. Mechanistically, we find that the protective action by Kv7.1 resides in a rapid translocation of channel proteins from intracellular stores to the plasma membrane, induced by adrenergic signaling. Gene silencing experiments targeting RAB GTPases, mediators of intracellular vesicle trafficking, showed that fast Kv7.1 recycling under acute stress conditions is RAB4A-dependent.Our data reveal a key mechanism underlying the rapid adaptation of human cardiomyocytes to adrenergic stress. These findings moreover aid to the understanding of disease pathology in long QT syndrome and bear important implications for safety pharmacological screening.
Collapse
Affiliation(s)
- Ilaria Piccini
- Department of Cardiovascular Medicine, Institute of Genetics of Heart Diseases, University of Münster Medical SchoolMünster, Germany.,Human Stem Cell Pluripotency Laboratory, Max Planck Institute for Molecular BiomedicineMünster, Germany
| | - Edda Fehrmann
- Institute of Pharmacology and Toxicology, University of MünsterMünster, Germany
| | - Stefan Frank
- Human Stem Cell Pluripotency Laboratory, Max Planck Institute for Molecular BiomedicineMünster, Germany.,Chemical Genomics Centre of the Max Planck SocietyDortmund, Germany
| | - Frank U Müller
- Institute of Pharmacology and Toxicology, University of MünsterMünster, Germany
| | - Boris Greber
- Human Stem Cell Pluripotency Laboratory, Max Planck Institute for Molecular BiomedicineMünster, Germany.,Chemical Genomics Centre of the Max Planck SocietyDortmund, Germany
| | - Guiscard Seebohm
- Department of Cardiovascular Medicine, Institute of Genetics of Heart Diseases, University of Münster Medical SchoolMünster, Germany
| |
Collapse
|
27
|
Bohnen MS, Peng G, Robey SH, Terrenoire C, Iyer V, Sampson KJ, Kass RS. Molecular Pathophysiology of Congenital Long QT Syndrome. Physiol Rev 2017; 97:89-134. [PMID: 27807201 PMCID: PMC5539372 DOI: 10.1152/physrev.00008.2016] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ion channels represent the molecular entities that give rise to the cardiac action potential, the fundamental cellular electrical event in the heart. The concerted function of these channels leads to normal cyclical excitation and resultant contraction of cardiac muscle. Research into cardiac ion channel regulation and mutations that underlie disease pathogenesis has greatly enhanced our knowledge of the causes and clinical management of cardiac arrhythmia. Here we review the molecular determinants, pathogenesis, and pharmacology of congenital Long QT Syndrome. We examine mechanisms of dysfunction associated with three critical cardiac currents that comprise the majority of congenital Long QT Syndrome cases: 1) IKs, the slow delayed rectifier current; 2) IKr, the rapid delayed rectifier current; and 3) INa, the voltage-dependent sodium current. Less common subtypes of congenital Long QT Syndrome affect other cardiac ionic currents that contribute to the dynamic nature of cardiac electrophysiology. Through the study of mutations that cause congenital Long QT Syndrome, the scientific community has advanced understanding of ion channel structure-function relationships, physiology, and pharmacological response to clinically employed and experimental pharmacological agents. Our understanding of congenital Long QT Syndrome continues to evolve rapidly and with great benefits: genotype-driven clinical management of the disease has improved patient care as precision medicine becomes even more a reality.
Collapse
Affiliation(s)
- M S Bohnen
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - G Peng
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - S H Robey
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - C Terrenoire
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - V Iyer
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - K J Sampson
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - R S Kass
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| |
Collapse
|
28
|
Zylla MM, Thomas D. Inherited Arrhythmias: Of Channels, Currents, and Swimming. Biophys J 2016; 110:1017-22. [PMID: 26958876 DOI: 10.1016/j.bpj.2016.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/07/2015] [Indexed: 02/06/2023] Open
Affiliation(s)
- Maura M Zylla
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
29
|
Margiotta A, Progida C, Bakke O, Bucci C. Rab7a regulates cell migration through Rac1 and vimentin. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:367-381. [PMID: 27888097 DOI: 10.1016/j.bbamcr.2016.11.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 11/09/2016] [Accepted: 11/19/2016] [Indexed: 01/17/2023]
Abstract
Rab7a, a small GTPase of the Rab family, is localized to late endosomes and controls late endocytic trafficking. The discovery of several Rab7a interacting proteins revealed that Rab7a function is closely connected to cytoskeletal elements. Indeed, Rab7a recruits on vesicles RILP and FYCO that are responsible for the movement of Rab7a-positive vesicles and/or organelles on microtubule tracks, but also directly interacts with Rac1, a fundamental regulator of actin cytoskeleton, and with peripherin and vimentin, two intermediate filament proteins. Considering all these interactions and, in particular, the fact that Rac1 and vimentin are key factors for cellular motility, we investigated a possible role of Rab7a in cell migration. We show here that Rab7a is needed for cell migration as Rab7a depletion causes slower migration of NCI H1299 cells affecting cell velocity and directness. Rab7a depletion negatively affects adhesion and spreading onto fibronectin substrates, altering β1-integrin activation, localization and intracellular trafficking, and myosin X localization. In fact, Rab7a-depleted cells show 40% less filopodia and active integrin accumulates at the leading edge of migrating cells. Furthermore, Rab7a depletion decreases the amount of active Rac1 but not its abundance and reduces the number of cells with vimentin filaments facing the wound, indicating that Rab7a has a role in the orientation of vimentin filaments during migration. In conclusion, our results demonstrate a key role of Rab7a in the regulation of different aspects of cell migration.
Collapse
Affiliation(s)
- Azzurra Margiotta
- Department of Biological and Environmental Sciences and Technologies, (DiSTeBA) University of Salento, Via Provinciale Monteroni 165, 73100 Lecce, Italy; Department of Biosciences, Centre for Immune Regulation, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Cinzia Progida
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Oddmund Bakke
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, (DiSTeBA) University of Salento, Via Provinciale Monteroni 165, 73100 Lecce, Italy.
| |
Collapse
|
30
|
Multiple Roles of the Small GTPase Rab7. Cells 2016; 5:cells5030034. [PMID: 27548222 PMCID: PMC5040976 DOI: 10.3390/cells5030034] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/11/2016] [Accepted: 08/12/2016] [Indexed: 12/16/2022] Open
Abstract
Rab7 is a small GTPase that belongs to the Rab family and controls transport to late endocytic compartments such as late endosomes and lysosomes. The mechanism of action of Rab7 in the late endocytic pathway has been extensively studied. Rab7 is fundamental for lysosomal biogenesis, positioning and functions, and for trafficking and degradation of several signaling receptors, thus also having implications on signal transduction. Several Rab7 interacting proteins have being identified leading to the discovery of a number of different important functions, beside its established role in endocytosis. Furthermore, Rab7 has specific functions in neurons. This review highlights and discusses the role and the importance of Rab7 on different cellular pathways and processes.
Collapse
|
31
|
Abstract
Cardiac delayed rectifier potassium channels conduct outward potassium currents during the plateau phase of action potentials and play pivotal roles in cardiac repolarization. These include IKs, IKr and the atrial specific IKur channels. In this article, we will review their molecular identities and biophysical properties. Mutations in the genes encoding delayed rectifiers lead to loss- or gain-of-function phenotypes, disrupt normal cardiac repolarization and result in various cardiac rhythm disorders, including congenital Long QT Syndrome, Short QT Syndrome and familial atrial fibrillation. We will also discuss the prospect of using delayed rectifier channels as therapeutic targets to manage cardiac arrhythmia.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmacology, College of Physicians & Surgeons of Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Kevin J Sampson
- Department of Pharmacology, College of Physicians & Surgeons of Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Robert S Kass
- Department of Pharmacology, College of Physicians & Surgeons of Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
32
|
Refaat MM, El Hage L, Steffensen AB, Hotait M, Schmitt N, Scheinman M, Badhwar N. Iron Overload Leading to Torsades de Pointes in β-Thalassemia and Long QT Syndrome. Card Electrophysiol Clin 2016; 8:247-256. [PMID: 26920202 DOI: 10.1016/j.ccep.2015.10.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The authors present a unique case of torsades de pointes in a β-thalassemia patient with early iron overload in the absence of any structural abnormalities as seen in hemochromatosis. Genetic testing showed a novel KCNQ1 gene mutation 1591C>T [Gln531Ter(X)]. Testing of the gene mutation in Xenopus laevis oocytes showed loss of function of the IKs current. The authors hypothesize that iron overload combined with the KCNQ1 gene mutation leads to prolongation of QTc and torsades de pointes.
Collapse
Affiliation(s)
- Marwan M Refaat
- Cardiology Division, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, American University of Beirut Medical Center, Beirut, Lebanon; Cardiac Electrophysiology, Cardiology, Department of Internal Medicine, American University of Beirut Faculty of Medicine and Medical Center, 3 Dag Hammarskjold Plaza, 8th Floor, New York, NY 10017, USA; Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center, 3 Dag Hammarskjold Plaza, 8th Floor, New York, NY 10017, USA
| | - Lea El Hage
- Division of Cardiology, Department of Medicine, University of California San Francisco Medical Center, 500 Parnassus Avenue, MUE-431, San Francisco, CA 94143-1354, USA
| | - Annette Buur Steffensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
| | - Mostafa Hotait
- Cardiology Division, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nicole Schmitt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
| | - Melvin Scheinman
- Division of Cardiology, Department of Medicine, University of California San Francisco Medical Center, 500 Parnassus Avenue, MUE-431, San Francisco, CA 94143-1354, USA
| | - Nitish Badhwar
- Division of Cardiology, Department of Medicine, University of California San Francisco Medical Center, 500 Parnassus Avenue, MUE-431, San Francisco, CA 94143-1354, USA.
| |
Collapse
|
33
|
Ohya S, Kito H, Hatano N, Muraki K. Recent advances in therapeutic strategies that focus on the regulation of ion channel expression. Pharmacol Ther 2016; 160:11-43. [PMID: 26896566 DOI: 10.1016/j.pharmthera.2016.02.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A number of different ion channel types are involved in cell signaling networks, and homeostatic regulatory mechanisms contribute to the control of ion channel expression. Profiling of global gene expression using microarray technology has recently provided novel insights into the molecular mechanisms underlying the homeostatic and pathological control of ion channel expression. It has demonstrated that the dysregulation of ion channel expression is associated with the pathogenesis of neural, cardiovascular, and immune diseases as well as cancers. In addition to the transcriptional, translational, and post-translational regulation of ion channels, potentially important evidence on the mechanisms controlling ion channel expression has recently been accumulated. The regulation of alternative pre-mRNA splicing is therefore a novel therapeutic strategy for the treatment of dominant-negative splicing disorders. Epigenetic modification plays a key role in various pathological conditions through the regulation of pluripotency genes. Inhibitors of pre-mRNA splicing and histone deacetyalase/methyltransferase have potential as potent therapeutic drugs for cancers and autoimmune and inflammatory diseases. Moreover, membrane-anchoring proteins, lysosomal and proteasomal degradation-related molecules, auxiliary subunits, and pharmacological agents alter the protein folding, membrane trafficking, and post-translational modifications of ion channels, and are linked to expression-defect channelopathies. In this review, we focused on recent insights into the transcriptional, spliceosomal, epigenetic, and proteasomal regulation of ion channel expression: Ca(2+) channels (TRPC/TRPV/TRPM/TRPA/Orai), K(+) channels (voltage-gated, KV/Ca(2+)-activated, KCa/two-pore domain, K2P/inward-rectifier, Kir), and Ca(2+)-activated Cl(-) channels (TMEM16A/TMEM16B). Furthermore, this review highlights expression of these ion channels in expression-defect channelopathies.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Hiroaki Kito
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Noriyuki Hatano
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya 464-8650, Japan
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya 464-8650, Japan.
| |
Collapse
|
34
|
Almilaji A, Pakladok T, Muñoz C, Elvira B, Sopjani M, Lang F. Upregulation of KCNQ1/KCNE1 K+ channels by Klotho. Channels (Austin) 2015; 8:222-9. [PMID: 24457979 DOI: 10.4161/chan.27662] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Klotho is a transmembrane protein expressed primarily in kidney, parathyroid gland, and choroid plexus. The extracellular domain could be cleaved off and released into the systemic circulation. Klotho is in part effective as β-glucuronidase regulating protein stability in the cell membrane. Klotho is a major determinant of aging and life span.Overexpression of Klotho increases and Klotho deficiency decreases life span. Klotho deficiency may further result in hearing loss and cardiac arrhythmia. The present study explored whether Klotho modifies activity and protein abundance of KCNQ1/KCNE1, a K(+) channel required for proper hearing and cardiac repolarization. To this end, cRNA encoding KCNQ1/KCNE1 was injected in Xenopus oocytes with or without additional injection of cRNA encoding Klotho. KCNQ1/KCNE1 expressing oocytes were treated with human recombinant Klotho protein (30 ng/mL) for 24 h. Moreover, oocytes which express both KCNQ1/KCNE1 and Klotho were treated with 10 μM DSA L (D-saccharic acid-1,4-lactone), a β-glucuronidase inhibitor. The KCNQ1/KCNE1 depolarization-induced current (I(Ks)) was determined utilizing dual electrode voltage clamp, while KCNQ1/KCNE1 protein abundance in the cell membrane was visualized utilizing specific antibody binding and quantified by chemiluminescence. KCNQ1/KCNE1 channel activity and KCNQ1/KCNE1 protein abundance were upregulated by coexpression of Klotho. The effect was mimicked by treatment with human recombinant Klotho protein (30 ng/mL) and inhibited by DSA L (10 μM). In conclusion, Klotho upregulates KCNQ1/KCNE1 channel activity by “mainly” enhancing channel protein abundance in the plasma cell membrane, an effect at least partially mediated through the β-glucuronidase activity of Klotho protein.
Collapse
|
35
|
Ballou LM, Lin RZ, Cohen IS. Control of cardiac repolarization by phosphoinositide 3-kinase signaling to ion channels. Circ Res 2015; 116:127-37. [PMID: 25552692 DOI: 10.1161/circresaha.116.303975] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Upregulation of phosphoinositide 3-kinase (PI3K) signaling is a common alteration in human cancer, and numerous drugs that target this pathway have been developed for cancer treatment. However, recent studies have implicated inhibition of the PI3K signaling pathway as the cause of a drug-induced long-QT syndrome in which alterations in several ion currents contribute to arrhythmogenic drug activity. Surprisingly, some drugs that were thought to induce long-QT syndrome by direct block of the rapid delayed rectifier (IKr) also seem to inhibit PI3K signaling, an effect that may contribute to their arrhythmogenicity. The importance of PI3K in regulating cardiac repolarization is underscored by evidence that QT interval prolongation in diabetes mellitus also may result from changes in multiple currents because of decreased insulin activation of PI3K in the heart. How PI3K signaling regulates ion channels to control the cardiac action potential is poorly understood. Hence, this review summarizes what is known about the effect of PI3K and its downstream effectors, including Akt, on sodium, potassium, and calcium currents in cardiac myocytes. We also refer to some studies in noncardiac cells that provide insight into potential mechanisms of ion channel regulation by this signaling pathway in the heart. Drug development and safety could be improved with a better understanding of the mechanisms by which PI3K regulates cardiac ion channels and the extent to which PI3K inhibition contributes to arrhythmogenic susceptibility.
Collapse
Affiliation(s)
- Lisa M Ballou
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.)
| | - Richard Z Lin
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.).
| | - Ira S Cohen
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.).
| |
Collapse
|
36
|
Recessive cardiac phenotypes in induced pluripotent stem cell models of Jervell and Lange-Nielsen syndrome: disease mechanisms and pharmacological rescue. Proc Natl Acad Sci U S A 2014; 111:E5383-92. [PMID: 25453094 DOI: 10.1073/pnas.1419553111] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Jervell and Lange-Nielsen syndrome (JLNS) is one of the most severe life-threatening cardiac arrhythmias. Patients display delayed cardiac repolarization, associated high risk of sudden death due to ventricular tachycardia, and congenital bilateral deafness. In contrast to the autosomal dominant forms of long QT syndrome, JLNS is a recessive trait, resulting from homozygous (or compound heterozygous) mutations in KCNQ1 or KCNE1. These genes encode the α and β subunits, respectively, of the ion channel conducting the slow component of the delayed rectifier K(+) current, IKs. We used complementary approaches, reprogramming patient cells and genetic engineering, to generate human induced pluripotent stem cell (hiPSC) models of JLNS, covering splice site (c.478-2A>T) and missense (c.1781G>A) mutations, the two major classes of JLNS-causing defects in KCNQ1. Electrophysiological comparison of hiPSC-derived cardiomyocytes (CMs) from homozygous JLNS, heterozygous, and wild-type lines recapitulated the typical and severe features of JLNS, including pronounced action and field potential prolongation and severe reduction or absence of IKs. We show that this phenotype had distinct underlying molecular mechanisms in the two sets of cell lines: the previously unidentified c.478-2A>T mutation was amorphic and gave rise to a strictly recessive phenotype in JLNS-CMs, whereas the missense c.1781G>A lesion caused a gene dosage-dependent channel reduction at the cell membrane. Moreover, adrenergic stimulation caused action potential prolongation specifically in JLNS-CMs. Furthermore, sensitivity to proarrhythmic drugs was strongly enhanced in JLNS-CMs but could be pharmacologically corrected. Our data provide mechanistic insight into distinct classes of JLNS-causing mutations and demonstrate the potential of hiPSC-CMs in drug evaluation.
Collapse
|
37
|
Abstract
Ion channels are essential for basic cellular function and for processes including sensory perception and intercellular communication in multicellular organisms. Voltage-gated potassium (Kv) channels facilitate dynamic cellular repolarization during an action potential, opening in response to membrane depolarization to facilitate K+ efflux. In both excitable and nonexcitable cells other, constitutively active, K+ channels provide a relatively constant repolarizing force to control membrane potential, ion homeostasis, and secretory processes. Of the forty known human Kv channel pore-forming α subunits that coassemble in various combinations to form the fundamental tetrameric channel pore and voltage sensor module, KCNQ1 is unique. KCNQ1 stands alone in having the capacity to form either channels that are voltage-dependent and require membrane depolarization for activation, or constitutively active channels. In mammals, KCNQ1 regulates processes including gastric acid secretion, thyroid hormone biosynthesis, salt and glucose homeostasis, and cell volume and in some species is required for rhythmic beating of the heart. In this review, the author discusses the unique functional properties, regulation, cell biology, diverse physiological roles, and involvement in human disease states of this chameleonic K+ channel.
Collapse
|
38
|
Steinke K, Sachse F, Ettischer N, Strutz‐Seebohm N, Henrion U, Rohrbeck M, Klosowski R, Wolters D, Brunner S, Franz W, Pott L, Munoz C, Kandolf R, Schulze‐Bahr E, Lang F, Klingel K, Seebohm G. Coxsackievirus B3 modulates cardiac ion channels. FASEB J 2013; 27:4108-21. [DOI: 10.1096/fj.13-230193] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Katja Steinke
- Institute for Genetics of Heart Diseases (IfGH)Department of Cardiovascular MedicineUniversity Hospital MünsterMünsterGermany
- Department of Biochemistry ICation Channel GroupRuhr University BochumBochumGermany
| | - Frank Sachse
- Nora Eccles Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUtahUSA
- Department of BioengineeringUniversity of UtahSalt Lake CityUtahUSA
| | - Nicole Ettischer
- Department of Molecular PathologyUniversity Hospital of TuebingenTuebingenGermany
| | - Nathalie Strutz‐Seebohm
- Institute for Genetics of Heart Diseases (IfGH)Department of Cardiovascular MedicineUniversity Hospital MünsterMünsterGermany
| | - Ulrike Henrion
- Institute for Genetics of Heart Diseases (IfGH)Department of Cardiovascular MedicineUniversity Hospital MünsterMünsterGermany
- Department of Physiology IUniversity of TuebingenTuebingenGermany
| | - Matthias Rohrbeck
- Institute for Genetics of Heart Diseases (IfGH)Department of Cardiovascular MedicineUniversity Hospital MünsterMünsterGermany
| | - Rafael Klosowski
- Department of Analytical ChemistryRuhr University BochumBochumGermany
| | - Dirk Wolters
- Department of Analytical ChemistryRuhr University BochumBochumGermany
| | - Stefan Brunner
- Department of Biochemistry ICation Channel GroupRuhr University BochumBochumGermany
| | - Wolfgang‐Michael Franz
- Department of Internal Medicine ILudwig Maximilians UniversityCampus GrosshadernMunichGermany
| | - Lutz Pott
- Department of Cellular PhysiologyRuhr University BochumBochumGermany
| | - Carlos Munoz
- Department of Physiology IUniversity of TuebingenTuebingenGermany
| | - Reinhard Kandolf
- Nora Eccles Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUtahUSA
- Department of BioengineeringUniversity of UtahSalt Lake CityUtahUSA
| | - Eric Schulze‐Bahr
- Institute for Genetics of Heart Diseases (IfGH)Department of Cardiovascular MedicineUniversity Hospital MünsterMünsterGermany
| | - Florian Lang
- Department of Physiology IUniversity of TuebingenTuebingenGermany
| | - Karin Klingel
- Department of Molecular PathologyUniversity Hospital of TuebingenTuebingenGermany
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH)Department of Cardiovascular MedicineUniversity Hospital MünsterMünsterGermany
| |
Collapse
|
39
|
Vandenberg LN, Morrie RD, Seebohm G, Lemire JM, Levin M. Rab GTPases are required for early orientation of the left-right axis in Xenopus. Mech Dev 2013; 130:254-71. [PMID: 23354119 PMCID: PMC10676213 DOI: 10.1016/j.mod.2012.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 11/14/2012] [Accepted: 11/16/2012] [Indexed: 02/04/2023]
Abstract
The earliest steps of left-right (LR) patterning in Xenopus embryos are driven by biased intracellular transport that ensures a consistently asymmetric localization of maternal ion channels and pumps in the first 2-4 blastomeres. The subsequent differential net efflux of ions by these transporters generates a bioelectrical asymmetry; this LR voltage gradient redistributes small signaling molecules along the LR axis that later regulate transcription of the normally left-sided Nodal. This system thus amplifies single cell chirality into a true left-right asymmetry across multi-cellular fields. Studies using molecular-genetic gain- and loss-of-function reagents have characterized many of the steps involved in this early pathway in Xenopus. Yet one key question remains: how is the chiral cytoskeletal architecture interpreted to localize ion transporters to the left or right side? Because Rab GTPases regulate nearly all aspects of membrane trafficking, we hypothesized that one or more Rab proteins were responsible for the directed, asymmetric shuttling of maternal ion channel or pump proteins. After performing a screen using dominant negative and wildtype (overexpressing) mRNAs for four different Rabs, we found that alterations in Rab11 expression randomize both asymmetric gene expression and organ situs. We also demonstrated that the asymmetric localization of two ion transporter subunits requires Rab11 function, and that Rab11 is closely associated with at least one of these subunits. Yet, importantly, we found that endogenous Rab11 mRNA and protein are expressed symmetrically in the early embryo. We conclude that Rab11-mediated transport is responsible for the movement of cargo within early blastomeres, and that Rab11 expression is required throughout the early embryo for proper LR patterning.
Collapse
Affiliation(s)
- Laura N. Vandenberg
- Center for Regenerative and Developmental Biology, and Biology Department, Tufts University Medford, MA 02155, United States
| | - Ryan D. Morrie
- Center for Regenerative and Developmental Biology, and Biology Department, Tufts University Medford, MA 02155, United States
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, Myocellular Electophysiology Group, University Hospital Münster, D-48149 Münster, Germany
| | - Joan M. Lemire
- Center for Regenerative and Developmental Biology, and Biology Department, Tufts University Medford, MA 02155, United States
| | - Michael Levin
- Center for Regenerative and Developmental Biology, and Biology Department, Tufts University Medford, MA 02155, United States
| |
Collapse
|
40
|
Lang F, Voelkl J. Therapeutic potential of serum and glucocorticoid inducible kinase inhibition. Expert Opin Investig Drugs 2013; 22:701-14. [PMID: 23506284 DOI: 10.1517/13543784.2013.778971] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Expression of serum-and-glucocorticoid-inducible kinase-1 (SGK1) is low in most cells, but dramatically increases under certain pathophysiological conditions, such as glucocorticoid or mineralocorticoid excess, inflammation with TGFβ release, hyperglycemia, cell shrinkage and ischemia. SGK1 is activated by insulin and growth factors via phosphatidylinositide-3-kinase, 3-phosphoinositide-dependent kinase and mammalian target of rapamycin. SGK1 sensitive functions include activation of ion channels (including epithelial Na(+) channel ENaC, voltage gated Na(+) channel SCN5A transient receptor potential channels TRPV4 - 6, Ca(2+) release activated Ca(2+) channel Orai1/STIM1, renal outer medullary K(+) channel ROMK, voltage gated K(+) channels KCNE1/KCNQ1, kainate receptor GluR6, cystic fibrosis transmembrane regulator CFTR), carriers (including Na(+),Cl(-) symport NCC, Na(+),K(+),2Cl(-) symport NKCC, Na(+)/H(+) exchangers NHE1 and NHE3, Na(+), glucose symport SGLT1, several amino acid transporters), and Na(+)/K(+)-ATPase. SGK1 regulates several enzymes (e.g., glycogen synthase kinase-3, ubiquitin-ligase Nedd4-2) and transcription factors (e.g., forkhead transcription factor 3a, β-catenin, nuclear factor kappa B). AREAS COVERED The phenotype of SGK1 knockout mice is mild and SGK1 is apparently dispensible for basic functions. Excessive SGK1 expression and activity, however, contributes to the pathophysiology of several disorders, including hypertension, obesity, diabetes, thrombosis, stroke, fibrosing disease, infertility and tumor growth. A SGK1 gene variant (prevalence ∼ 3 - 5% in Caucasians and ∼ 10% in Africans) is associated with hypertension, stroke, obesity and type 2 diabetes. SGK1 inhibitors have been developed and shown to reduce blood pressure of hyperinsulinemic mice and to counteract tumor cell survival. EXPERT OPINION Targeting SGK1 may be a therapeutic option in several clinical conditions, including metabolic syndrome and tumor growth.
Collapse
Affiliation(s)
- Florian Lang
- University of Tuebingen, Department of Physiology, Tuebingen, Germany.
| | | |
Collapse
|
41
|
Nguyen N, Kozer-Gorevich N, Gliddon BL, Smolka AJ, Clayton AH, Gleeson PA, van Driel IR. Independent trafficking of the KCNQ1 K+ channel and H+-K+-ATPase in gastric parietal cells from mice. Am J Physiol Gastrointest Liver Physiol 2013; 304:G157-66. [PMID: 23154976 DOI: 10.1152/ajpgi.00346.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastric acid secretion by the H(+)-K(+)-ATPase at the apical surface of activated parietal cells requires luminal K(+) provided by the KCNQ1/KCNE2 K(+) channel. However, little is known about the trafficking and relative spatial distribution of KCNQ1 and H(+)-K(+)-ATPase in resting and activated parietal cells and the capacity of KCNQ1 to control acid secretion. Here we show that inhibition of KCNQ1 activity quickly curtails gastric acid secretion in vivo, even when the H(+)-K(+)-ATPase is permanently anchored in the apical membrane, demonstrating a key role of the K(+) channel in controlling acid secretion. Three-dimensional imaging analysis of isolated mouse gastric units revealed that the majority of KCNQ1 resides in an intracytoplasmic, Rab11-positive compartment in resting parietal cells, distinct from H(+)-K(+)-ATPase-enriched tubulovesicles. Upon activation, there was a significant redistribution of H(+)-K(+)-ATPase and KCNQ1 from intracytoplasmic compartments to the apical secretory canaliculi. Significantly, high Förster resonance energy transfer was detected between H(+)-K(+)-ATPase and KCNQ1 in activated, but not resting, parietal cells. These findings demonstrate that H(+)-K(+)-ATPase and KCNQ1 reside in independent intracytoplasmic membrane compartments, or membrane domains, and upon activation of parietal cells, both membrane proteins are transported, possibly via Rab11-positive recycling endosomes, to apical membranes, where the two molecules are closely physically opposed. In addition, these studies indicate that acid secretion is regulated by independent trafficking of KCNQ1 and H(+)-K(+)-ATPase.
Collapse
Affiliation(s)
- Nhung Nguyen
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Australia
| | | | | | | | | | | | | |
Collapse
|
42
|
Lang F, Shumilina E. Regulation of ion channels by the serum- and glucocorticoid-inducible kinase SGK1. FASEB J 2012; 27:3-12. [PMID: 23012321 DOI: 10.1096/fj.12-218230] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ubiquitously expressed serum- and glucocorticoid-inducible kinase-1 (SGK1) is genomically regulated by cell stress (including cell shrinkage) and several hormones (including gluco- and mineralocorticoids). SGK1 is activated by insulin and growth factors through PI3K and 3-phosphoinositide-dependent kinase PDK1. SGK1 activates a wide variety of ion channels (e.g., ENaC, SCN5A, TRPV4-6, ROMK, Kv1.3, Kv1.5, Kv4.3, KCNE1/KCNQ1, KCNQ4, ASIC1, GluR6, ClCKa/barttin, ClC2, CFTR, and Orai/STIM), which participate in the regulation of transport, hormone release, neuroexcitability, inflammation, cell proliferation, and apoptosis. SGK1-sensitive ion channels participate in the regulation of renal Na(+) retention and K(+) elimination, blood pressure, gastric acid secretion, cardiac action potential, hemostasis, and neuroexcitability. A common (∼3-5% prevalence in Caucasians and ∼10% in Africans) SGK1 gene variant is associated with increased blood pressure and body weight as well as increased prevalence of type II diabetes and stroke. SGK1 further contributes to the pathophysiology of allergy, peptic ulcer, fibrosing disease, ischemia, tumor growth, and neurodegeneration. The effect of SGK1 on channel activity is modest, and the channels do not require SGK1 for basic function. SGK1-dependent ion channel regulation may thus become pathophysiologically relevant primarily after excessive (pathological) expression. Therefore, SGK1 may be considered an attractive therapeutic target despite its broad range of functions.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, University of Tuebingen, Gmelinstrasse 5, 72076 Tuebingen, Germany.
| | | |
Collapse
|
43
|
Kanda VA, Abbott GW. KCNE Regulation of K(+) Channel Trafficking - a Sisyphean Task? Front Physiol 2012; 3:231. [PMID: 22754540 PMCID: PMC3385356 DOI: 10.3389/fphys.2012.00231] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 06/08/2012] [Indexed: 11/16/2022] Open
Abstract
Voltage-gated potassium (Kv) channels shape the action potentials of excitable cells and regulate membrane potential and ion homeostasis in excitable and non-excitable cells. With 40 known members in the human genome and a variety of homomeric and heteromeric pore-forming α subunit interactions, post-translational modifications, cellular locations, and expression patterns, the functional repertoire of the Kv α subunit family is monumental. This versatility is amplified by a host of interacting proteins, including the single membrane-spanning KCNE ancillary subunits. Here, examining both the secretory and the endocytic pathways, we review recent findings illustrating the surprising virtuosity of the KCNE proteins in orchestrating not just the function, but also the composition, diaspora and retrieval of channels formed by their Kv α subunit partners.
Collapse
Affiliation(s)
- Vikram A Kanda
- Department of Biology, Manhattan College Riverdale, New York, NY, USA
| | | |
Collapse
|
44
|
Wilmes J, Haddad-Tóvolli R, Alesutan I, Munoz C, Sopjani M, Pelzl L, Bogatikov E, Fedele G, Faggio C, Seebohm G, Föller M, Lang F. Regulation of KCNQ1/KCNE1 by β-catenin. Mol Membr Biol 2012; 29:87-94. [PMID: 22583083 DOI: 10.3109/09687688.2012.678017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
β-catenin, a multifunctional protein expressed in all tissues including the heart stimulates the expression of several genes important for cell proliferation. Signaling involving ß-catenin participates in directing cardiac development and in the pathophysiology of cardiac hypertrophy. Nothing is known, however, on the role of β-catenin in the regulation of cardiac ion channels. The present study explored the functional interaction of β-catenin and KCNE1/KCNQ1, the K⁺ channel complex underlying the slowly activating outwardly rectifying K⁺ current. To this end, KCNE1/KCNQ1 was expressed in Xenopus oocytes with and without β-catenin and the depolarization (up to + 80 mV) induced current (I(Ks)) was determined using the two-electrode voltage clamp. As a result, β-catenin enhanced I(Ks) by 30%. The effect of β-catenin on I(Ks) was not affected by actinomycin D (10 μM), an inhibitor of transcription, indicating that β-catenin was not effective as transcription factor. Confocal microscopy revealed that β-catenin enhanced the KCNE1/KCNQ1 protein abundance in the cell membrane. Exposure of the oocytes to brefeldin A (5 μM), an inhibitor of vesicle insertion, was followed by a decline of I(Ks), which was then similar in oocytes expressing KCNE1/KCNQ1 together with β-catenin and in oocytes expressing KCNE1/KCNQ1 alone. In conclusion, β-catenin enhances I(Ks) by increasing the KCNE1/KCNQ1 protein abundance in the cell membrane, an effect requiring vesicle insertion into the cell membrane.
Collapse
Affiliation(s)
- Jan Wilmes
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Roura-Ferrer M, Solé L, Oliveras A, Villarroel A, Comes N, Felipe A. Targeting of Kv7.5 (KCNQ5)/KCNE channels to surface microdomains of cell membranes. Muscle Nerve 2012; 45:48-54. [PMID: 22190306 DOI: 10.1002/mus.22231] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Kv7.5 (KCNQ5) channels conduct M-type potassium currents in the brain, are expressed in skeletal muscle, and contribute to vascular muscle tone. METHODS We coexpressed Kv7.5 and KCNE1-3 peptides in HEK293 cells and then analyzed their association using electrophysiology and co-immunoprecipitation, assessed localization using confocal microscopy, examined targeting of the oligomeric channels to cholesterol-rich membrane surface microdomains using lipid raft isolation, and evaluated their membrane dynamics using fluorescence recovery after photobleaching (FRAP). RESULTS Kv7.5 forms oligomeric channels specifically with KCNE1 and KCNE3. The expression of Kv7.5 targeted to cholesterol-rich membrane surface microdomains was very low. Oligomeric Kv7.5/KCNE1 and Kv7.5/KCNE3 channels did not localize to lipid rafts. However, Kv7.5 association impaired KCNE3 expression in lipid raft microdomains. CONCLUSIONS Our results indicate that Kv7.5 contributes to the spatial regulation of KCNE3. This new scenario could greatly assist in determining the physiological relevance of putative KCNE3 interactions in nerve and muscle.
Collapse
Affiliation(s)
- Meritxell Roura-Ferrer
- Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona, Avenida Diagonal 645, E-08028 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
46
|
Seebohm G, Strutz-Seebohm N, Ursu ON, Preisig-Müller R, Zuzarte M, Hill EV, Kienitz MC, Bendahhou S, Fauler M, Tapken D, Decher N, Collins A, Jurkat-Rott K, Steinmeyer K, Lehmann-Horn F, Daut J, Tavaré JM, Pott L, Bloch W, Lang F. Altered stress stimulation of inward rectifier potassium channels in Andersen-Tawil syndrome. FASEB J 2011; 26:513-22. [PMID: 22002906 DOI: 10.1096/fj.11-189126] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Inward rectifier potassium channels of the Kir2 subfamily are important determinants of the electrical activity of brain and muscle cells. Genetic mutations in Kir2.1 associate with Andersen-Tawil syndrome (ATS), a familial disorder leading to stress-triggered periodic paralysis and ventricular arrhythmia. To identify the molecular mechanisms of this stress trigger, we analyze Kir channel function and localization electrophysiologically and by time-resolved confocal microscopy. Furthermore, we employ a mathematical model of muscular membrane potential. We identify a novel corticoid signaling pathway that, when activated by glucocorticoids, leads to enrichment of Kir2 channels in the plasma membranes of mammalian cell lines and isolated cardiac and skeletal muscle cells. We further demonstrate that activation of this pathway can either partly restore (40% of cases) or further impair (20% of cases) the function of mutant ATS channels, depending on the particular Kir2.1 mutation. This means that glucocorticoid treatment might either alleviate or deteriorate symptoms of ATS depending on the patient's individual Kir2.1 genotype. Thus, our findings provide a possible explanation for the contradictory effects of glucocorticoid treatment on symptoms in patients with ATS and may open new pathways for the design of personalized medicines in ATS therapy.
Collapse
Affiliation(s)
- Guiscard Seebohm
- Department of Biochemistry I-Cation Channel Group, Ruhr University Bochum, Bochum, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Neal AM, Taylor HC, Millar ID, Kibble JD, White SJ, Robson L. Renal defects in KCNE1 knockout mice are mimicked by chromanol 293B in vivo: identification of a KCNE1-regulated K+ conductance in the proximal tubule. J Physiol 2011; 589:3595-609. [PMID: 21576273 PMCID: PMC3167120 DOI: 10.1113/jphysiol.2011.209155] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Non-technical summary The kidney plays a critical role in regulating body fluid volume and blood pressure by conserving ions, solutes and water. Knowing the processes that underpin the handling of ions, solutes and water by the kidney is essential to our understanding of fluid and blood pressure regulation. Movement of ions is mediated by specific transport proteins found in the membranes of kidney cells. These proteins are regulated by additional proteins, called accessory proteins. In the current study, we have examined the role of the accessory protein KCNE1 in regulating a channel, KCNQ1, which is important in kidney function. We have observed that in the absence of KCNE1 the kidney has difficulty conserving sodium, chloride and water. However, by using specific inhibitors of these proteins we have also determined that although KCNE1 has a role in kidney function, the mechanism of its action is unlikely to be by regulating the protein KCNQ1. Abstract KCNE1 is a protein of low molecular mass that is known to regulate the chromanol 293B and clofilium-sensitive K+ channel, KCNQ1, in a number of tissues. Previous work on the kidney of KCNE1 and KCNQ1 knockout mice has revealed that these animals have different renal phenotypes, suggesting that KCNE1 may not regulate KCNQ1 in the renal system. In the current study, in vivo clearance approaches and whole cell voltage-clamp recordings from isolated renal proximal tubules were used to examine the physiological role of KCNE1. Data from wild-type mice were compared to those from KCNE1 knockout mice. In clearance studies the KCNE1 knockout mice had an increased fractional excretion of Na+, Cl−, HCO3− and water. This profile was mimicked in wild-type mice by infusion of chromanol 293B, while chromanol was without effect in KCNE1 knockout animals. Clofilium also increased the fractional excretion of Na+, Cl− and water, but this was observed in both wild-type and knockout mice, suggesting that KCNE1 was regulating a chromanol-sensitive but clofilium-insensitive pathway. In whole cell voltage clamp recordings from proximal tubules, a chromanol-sensitive, K+-selective conductance was identified that was absent in tubules from knockout animals. The properties of this conductance were not consistent with its being mediated by KCNQ1, suggesting that KCNE1 regulates another K+ channel in the renal proximal tubule. Taken together these data suggest that KCNE1 regulates a K+-selective conductance in the renal proximal tubule that plays a relatively minor role in driving the transport of Na+, Cl− and HCO3−.
Collapse
Affiliation(s)
- A M Neal
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | | | | | | | | | | |
Collapse
|
48
|
Umbach AT, Pathare G, Föller M, Brosens JJ, Artunc F, Lang F. SGK1-dependent salt appetite in pregnant mice. Acta Physiol (Oxf) 2011; 202:39-45. [PMID: 21251236 DOI: 10.1111/j.1748-1716.2011.02251.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM Pregnancy is typically paralleled by substantial increase in maternal extracellular fluid volume, requiring net accumulation of water and NaCl. The positive water and salt balance is accomplished at least in part by increased uptake of salt secondary to enhanced salt appetite. Little is known about the underlying cellular mechanisms. Stimulation of salt appetite by mineralocorticoids, however, is known to be dependent on the serum- and glucocorticoid-inducible kinase SGK1. METHODS To test for a role of SGK1 in the stimulation of salt appetite during pregnancy, fluid intake was recorded in pregnant SGK1 knockout mice (sgk1(-/-) ) and their wild type littermates (sgk1(+/+) ). The mice were offered two bottles, one with plain water and the other with isotonic saline. RESULTS In early pregnancy, i.e. up to 10 days prior to parturition, the sgk1(+/+) mice displayed a significant preference for saline, whereas the sgk1(-/-) mice preferred water. Accordingly, the water intake was significantly smaller and saline intake was significantly larger in sgk1(+/+) mice than in sgk1(-/-) mice and the preference for water was significantly stronger in sgk1(-/-) mice than in sgk1(+/+) mice. Plasma aldosterone levels were higher in sgk1(-/-) mice than in sgk1(+/+) mice, a difference contrasting the enhanced salt appetite of sgk1(+/+) mice. CONCLUSIONS SGK1 participates in the stimulation of salt appetite during pregnancy.
Collapse
Affiliation(s)
- A T Umbach
- Department of Physiology, University of Tübingen, Germany
| | | | | | | | | | | |
Collapse
|
49
|
Van Horn WD, Vanoye CG, Sanders CR. Working model for the structural basis for KCNE1 modulation of the KCNQ1 potassium channel. Curr Opin Struct Biol 2011; 21:283-91. [PMID: 21296569 DOI: 10.1016/j.sbi.2011.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 01/03/2011] [Accepted: 01/04/2011] [Indexed: 12/19/2022]
Abstract
The voltage-gated potassium channel KCNQ1 (Kv7.1) is modulated by KCNE1 (minK) to generate the I(Ks) current crucial to heartbeat. Defects in either protein result in serious cardiac arrhythmias. Recently developed structural models of the open and closed state KCNQ1/KCNE1 complexes offer a compelling explanation for how KCNE1 slows channel opening and provides a platform from which to refine and test hypotheses for other aspects of KCNE1 modulation. These working models were developed using an integrative approach based on results from nuclear magnetic resonance spectroscopy, electrophysiology, biochemistry, and computational methods-an approach that can be applied iteratively for model testing and revision. We present a critical review of these structural models, illustrating the strengths and challenges of the integrative approach.
Collapse
Affiliation(s)
- Wade D Van Horn
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-8725, USA
| | | | | |
Collapse
|
50
|
Alesutan I, Föller M, Sopjani M, Dërmaku-Sopjani M, Zelenak C, Fröhlich H, Velic A, Fraser S, Kemp BE, Seebohm G, Völkl H, Lang F. Inhibition of the heterotetrameric K+ channel KCNQ1/KCNE1 by the AMP-activated protein kinase. Mol Membr Biol 2011; 28:79-89. [PMID: 21231794 DOI: 10.3109/09687688.2010.520037] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The heterotetrameric K(+)-channel KCNQ1/KCNE1 is expressed in heart, skeletal muscle, liver and several epithelia including the renal proximal tubule. In the heart, it contributes to the repolarization of cardiomyocytes. The repolarization is impaired in ischemia. Ischemia stimulates the AMP-activated protein kinase (AMPK), a serine/threonine kinase, sensing energy depletion and stimulating several cellular mechanisms to enhance energy production and to limit energy utilization. AMPK has previously been shown to downregulate the epithelial Na(+) channel ENaC, an effect mediated by the ubiquitin ligase Nedd4-2. The present study explored whether AMPK regulates KCNQ1/KCNE1. To this end, cRNA encoding KCNQ1/KCNE1 was injected into Xenopus oocytes with and without additional injection of wild type AMPK (AMPKα1 + AMPKβ1 + AMPKγ1), of the constitutively active (γR70Q)AMPK (α1β1γ1(R70Q)), of the kinase dead mutant (αK45R)AMPK (α1(K45R)β1γ1), or of the ubiquitin ligase Nedd4-2. KCNQ1/KCNE1 activity was determined in two electrode voltage clamp experiments. Moreover, KCNQ1 abundance in the cell membrane was determined by immunostaining and subsequent confocal imaging. As a result, wild type and constitutively active AMPK significantly reduced KCNQ1/KCNE1-mediated currents and reduced KCNQ1 abundance in the cell membrane. Similarly, Nedd4-2 decreased KCNQ1/KCNE1-mediated currents and KCNQ1 protein abundance in the cell membrane. Activation of AMPK in isolated perfused proximal renal tubules by AICAR (10 mM) was followed by significant depolarization. In conclusion, AMPK is a potent regulator of KCNQ1/KCNE1.
Collapse
Affiliation(s)
- Ioana Alesutan
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|