1
|
Huo J, Mo L, Lv X, Du Y, Yang H. Ion Channel Regulation in Caveolae and Its Pathological Implications. Cells 2025; 14:631. [PMID: 40358155 PMCID: PMC12071496 DOI: 10.3390/cells14090631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Caveolae are distinctive, flask-shaped structures within the cell membrane that play critical roles in cellular signal transduction, ion homeostasis, and mechanosensation. These structures are composed of the caveolin protein family and are enriched in cholesterol and sphingolipids, creating a unique lipid microdomain. Caveolae contribute to the functional regulation of various ion channels through both physical interactions and involvement in complex signaling networks. Ion channels localized within caveolae are involved in critical cellular processes such as the generation and propagation of action potentials, cellular responses to mechanical forces, and regulation of metabolism. Dysregulation of caveolae function has been linked to the development of various diseases, including cardiovascular disorders, neurodegenerative diseases, metabolic syndrome, and cancer. This review summarizes the ion channel function and regulation in caveolae, and their pathological implications, offering new insights into their potential as therapeutic targets for ion channel-related diseases.
Collapse
Affiliation(s)
| | | | | | | | - Huaqian Yang
- Department of Cardiology, The Fourth Affiliated Hospital, Cyrus Tang Medical Institute, Medical College, Soochow University, Suzhou 215028, China; (L.M.); (X.L.); (Y.D.)
| |
Collapse
|
2
|
Zhuang W, Mun SY, Park WS. Direct effects of antipsychotics on potassium channels. Biochem Biophys Res Commun 2025; 749:151344. [PMID: 39842331 DOI: 10.1016/j.bbrc.2025.151344] [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: 11/19/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Schizophrenia (SCZ) and bipolar disorder (BD) and are severe psychiatric conditions that contribute to disability and increased healthcare costs globally. Although first-, second-, and third-generation antipsychotics are available for treating BD and SCZ, most have various side effects unrelated to their unique functions. Many antipsychotics affect K+ channels (Kv, KCa, Kir, K2P, and other channels), which change the functions of various organs. This review summarizes the biological actions of antipsychotics, including off-target side effects involving K+ channels.
Collapse
Affiliation(s)
- Wenwen Zhuang
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Seo-Yeong Mun
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea.
| |
Collapse
|
3
|
Kelly C, Kiltschewskij DJ, Leong AJW, Haw TJ, Croft AJ, Balachandran L, Chen D, Bond DR, Lee HJ, Cairns MJ, Sverdlov AL, Ngo DTM. Identifying common pathways for doxorubicin and carfilzomib-induced cardiotoxicities: transcriptomic and epigenetic profiling. Sci Rep 2025; 15:4395. [PMID: 39910168 PMCID: PMC11799237 DOI: 10.1038/s41598-025-87442-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
Cancer therapy-related cardiovascular toxicity (CTR-CVT) is now recognised as one of the leading causes of long-term morbidity and mortality in cancer patients. To date, potential overlapping cardiotoxicity mechanism(s) across different chemotherapeutic classes have not been elucidated. Doxorubicin, an anthracycline, and Carfilzomib, a proteasome inhibitor, are both known to cause heart failure in some patients. Given this common cardiotoxic effect of these chemotherapies, we aimed to investigate differential and common mechanism(s) associated with Doxorubicin and Carfilzomib-induced cardiac dysfunction. Primary human cardiomyocyte-like cells (HCM-ls) were treated with 1 µM of either Doxorubicin or Carfilzomib for 72 h. Both Doxorubicin and Carfilzomib induced a significant reduction in HCM cell viability and cell damage. DNA methylation analysis performed using MethylationEPIC array showed distinct and common changes induced by Doxorubicin and Carfilzomib (10,270 or approximately 12.9% of the DMPs for either treatment overlapped). RNA-seq analyses identified 5,643 differentially expressed genes (DEGs) that were commonly dysregulated for both treatments. Pathway analysis revealed that the PI3K-Akt signalling pathway was the most significantly enriched pathway with common DEGs, shared between Doxorubicin and Carfilzomib. We identified that there are shared cardiotoxicity mechanisms for Doxorubicin and Carfilzomib pathways that can be potential therapeutic targets for treatments across 2 classes of anti-cancer agents.
Collapse
Affiliation(s)
- Conagh Kelly
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
| | - Dylan J Kiltschewskij
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Angeline J W Leong
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Tatt Jhong Haw
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Amanda J Croft
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Lohis Balachandran
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Dongqing Chen
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
| | - Danielle R Bond
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Heather J Lee
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Aaron L Sverdlov
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia.
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia.
- Cardiovascular Department, John Hunter Hospital, New Lambton Heights, NSW, Australia.
| | - Doan T M Ngo
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia.
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia.
| |
Collapse
|
4
|
Tao Y, Yao K, Wu J, Xu T, Lin J, Qin Y, Su D, Cai S, Yu W, Chen X. Intravenous anesthetics have differential effects on human potassium channels. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1594-1603. [PMID: 40384046 PMCID: PMC11659785 DOI: 10.3724/abbs.2024151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/02/2024] [Indexed: 05/20/2025] Open
Abstract
General anesthetics are widely used in the clinic and greatly promote the development of surgery. However, the incidence of cardiovascular and respiratory complications caused by general anesthetics is still high, and the underlying mechanisms remain incompletely understood. Potassium channels are widely expressed in the heart and blood vessels and participate in regulating blood pressure, heart rate, and other physiological parameters. Whether they are directly affected by intravenous general anesthetics is unclear. Here, we independently express four classes of potassium channels, TASK-1, TASK-3, Kv1.5, Kv2.1, Kir2.1, SK1 and SK3, in Xenopus oocytes. The effects of propofol, pentobarbital and ketamine on these channels are evaluated by their current change. We find that propofol and ketamine potentiate TASK-3 and SK3 current respectively, while pentobarbital and ketamine inhibit SK1 current. To identify the key residues in TASK-3, SK1 and SK3 that interact with intravenous anesthetics, we predict homology models of the three channels and perform molecular docking simulations. The results show that propofol forms a hydrogen bond with Q126 of TASK-3, ketamine forms a hydrogen bond with S290 of SK1 and S467 of SK3, while pentobarbital forms hydrogen bonds with S330 and T358 of SK1. As these potassium channels are closely related to respiratory system regulation, cardiac rhythm and vasodilation, our study provides a new perspective for further study on the mechanism of general anesthetics-induced respiratory and circulatory side effects.
Collapse
Affiliation(s)
- Ying Tao
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Kejie Yao
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai 200031China; University of Chinese Academy of SciencesBeijing101408China
| | - Jing Wu
- Department of Equipment and MaterialsBiomedical R&D Project TeamZhongshan HospitalFudan UniversityShanghai200031China
| | - Tian Xu
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Junhui Lin
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Yi Qin
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Diansan Su
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Shiqing Cai
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai 200031China; University of Chinese Academy of SciencesBeijing101408China
| | - Weifeng Yu
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Xuemei Chen
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| |
Collapse
|
5
|
Stary-Weinzinger A. In silico models of the macromolecular Na V1.5-K IR2.1 complex. Front Physiol 2024; 15:1362964. [PMID: 38468705 PMCID: PMC10925717 DOI: 10.3389/fphys.2024.1362964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/07/2024] [Indexed: 03/13/2024] Open
Abstract
In cardiac cells, the expression of the cardiac voltage-gated Na+ channel (NaV1.5) is reciprocally regulated with the inward rectifying K+ channel (KIR2.1). These channels can form macromolecular complexes that pre-assemble early during forward trafficking (transport to the cell membrane). In this study, we present in silico 3D models of NaV1.5-KIR2.1, generated by rigid-body protein-protein docking programs and deep learning-based AlphaFold-Multimer software. Modeling revealed that the two channels could physically interact with each other along the entire transmembrane region. Structural mapping of disease-associated mutations revealed a hotspot at this interface with several trafficking-deficient variants in close proximity. Thus, examining the role of disease-causing variants is important not only in isolated channels but also in the context of macromolecular complexes. These findings may contribute to a better understanding of the life-threatening cardiovascular diseases underlying KIR2.1 and NaV1.5 malfunctions.
Collapse
Affiliation(s)
- Anna Stary-Weinzinger
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Struckman HL, Moise N, King DR, Soltisz A, Buxton A, Dunlap I, Chen Z, Radwański PB, Weinberg SH, Veeraraghavan R. Unraveling Impacts of Chamber-Specific Differences in Intercalated Disc Ultrastructure and Molecular Organization on Cardiac Conduction. JACC Clin Electrophysiol 2023; 9:2425-2443. [PMID: 37498248 PMCID: PMC11102000 DOI: 10.1016/j.jacep.2023.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Propagation of action potentials through the heart coordinates the heartbeat. Thus, intercalated discs, specialized cell-cell contact sites that provide electrical and mechanical coupling between cardiomyocytes, are an important target for study. Impaired propagation leads to arrhythmias in many pathologies, where intercalated disc remodeling is a common finding, hence the importance and urgency of understanding propagation dependence on intercalated disc structure. Conventional modeling approaches cannot predict changes in propagation elicited by perturbations that alter intercalated disc ultrastructure or molecular organization, because of lack of quantitative structural data at subcellular through nano scales. OBJECTIVES This study sought to quantify intercalated disc structure at these spatial scales in the healthy adult mouse heart and relate them to chamber-specific properties of propagation as a precursor to understanding the effects of pathological intercalated disc remodeling. METHODS Using super-resolution light microscopy, electron microscopy, and computational image analysis, we provide here the first ever systematic, multiscale quantification of intercalated disc ultrastructure and molecular organization. RESULTS By incorporating these data into a rule-based model of cardiac tissue with realistic intercalated disc structure, and comparing model predictions of electrical propagation with experimental measures of conduction velocity, we reveal that atrial intercalated discs can support faster conduction than their ventricular counterparts, which is normally masked by interchamber differences in myocyte geometry. Further, we identify key ultrastructural and molecular organization features underpinning the ability of atrial intercalated discs to support faster conduction. CONCLUSIONS These data provide the first stepping stone to elucidating chamber-specific effects of pathological intercalated disc remodeling, as occurs in many arrhythmic diseases.
Collapse
Affiliation(s)
- Heather L Struckman
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Nicolae Moise
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - D Ryan King
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Andrew Soltisz
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Andrew Buxton
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Izabella Dunlap
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Zhenhui Chen
- Krannert Cardiovascular Research Center, Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Przemysław B Radwański
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA; Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Seth H Weinberg
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.
| |
Collapse
|
7
|
Huo JY, Feng YL, Chen YT, Yang B, Zhi YT, Wang HJ, Yang HQ. Caveolin-3 negatively regulates endocytic recycling of cardiac K ATP channels. Am J Physiol Cell Physiol 2023; 325:C1106-C1118. [PMID: 37746698 DOI: 10.1152/ajpcell.00266.2023] [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: 06/19/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/26/2023]
Abstract
Sarcolemmal ATP-sensitive potassium (KATP) channels play a vital role in cardioprotection. Cardiac KATP channels are enriched in caveolae and physically interact with the caveolae structural protein caveolin-3 (Cav3). Disrupting caveolae impairs the regulation of KATP channels through several signaling pathways. However, the direct functional effect of Cav3 on KATP channels is still poorly understood. Here, we used the cardiac KATP channel subtype, Kir6.2/SUR2A, and showed that Cav3 greatly reduced KATP channel surface density and current amplitude in a caveolae-independent manner. A screen of Cav3 functional domains revealed that a 25 amino acid region in the membrane attachment domain of Cav3 is the minimal effective segment (MAD1). The peptide corresponding to the MAD1 segment decreased KATP channel current in a concentration-dependent manner with an IC50 of ∼5 μM. The MAD1 segment prevented KATP channel recycling, thus decreasing KATP channel surface density and abolishing the cardioprotective effect of ischemic preconditioning. Our research identified the Cav3 MAD1 segment as a novel negative regulator of KATP channel recycling, providing pharmacological potential in the treatment of diseases with KATP channel trafficking defects.NEW & NOTEWORTHY Cardiac KATP channels physically interact with caveolin-3 in caveolae. In this study, we investigated the functional effect of caveolin-3 on KATP channel activity and identified a novel segment (MAD1) in the C-terminus domain of Caveolin-3 that negatively regulates KATP channel surface density and current amplitude by impairing KATP channel recycling. The peptide corresponding to the MAD1 segment abolished the cardioprotective effect of ischemic preconditioning.
Collapse
Affiliation(s)
- Jian-Yi Huo
- Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Yu-Long Feng
- Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Yue-Tong Chen
- Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Bo Yang
- Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Ya-Ting Zhi
- Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Hao-Jie Wang
- Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Hua-Qian Yang
- Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| |
Collapse
|
8
|
Markandeya YS, Gregorich ZR, Feng L, Ramchandran V, O' Hara T, Vaidyanathan R, Mansfield C, Keefe AM, Beglinger CJ, Best JM, Kalscheur MM, Lea MR, Hacker TA, Gorelik J, Trayanova NA, Eckhardt LL, Makielski JC, Balijepalli RC, Kamp TJ. Caveolin-3 and Caveolae regulate ventricular repolarization. J Mol Cell Cardiol 2023; 177:38-49. [PMID: 36842733 PMCID: PMC10065933 DOI: 10.1016/j.yjmcc.2023.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 02/03/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
RATIONALE Flask-shaped invaginations of the cardiomyocyte sarcolemma called caveolae require the structural protein caveolin-3 (Cav-3) and host a variety of ion channels, transporters, and signaling molecules. Reduced Cav-3 expression has been reported in models of heart failure, and variants in CAV3 have been associated with the inherited long-QT arrhythmia syndrome. Yet, it remains unclear whether alterations in Cav-3 levels alone are sufficient to drive aberrant repolarization and increased arrhythmia risk. OBJECTIVE To determine the impact of cardiac-specific Cav-3 ablation on the electrophysiological properties of the adult mouse heart. METHODS AND RESULTS Cardiac-specific, inducible Cav3 homozygous knockout (Cav-3KO) mice demonstrated a marked reduction in Cav-3 expression by Western blot and loss of caveolae by electron microscopy. However, there was no change in macroscopic cardiac structure or contractile function. The QTc interval was increased in Cav-3KO mice, and there was an increased propensity for ventricular arrhythmias. Ventricular myocytes isolated from Cav-3KO mice exhibited a prolonged action potential duration (APD) that was due to reductions in outward potassium currents (Ito, Iss) and changes in inward currents including slowed inactivation of ICa,L and increased INa,L. Mathematical modeling demonstrated that the changes in the studied ionic currents were adequate to explain the prolongation of the mouse ventricular action potential. Results from human iPSC-derived cardiomyocytes showed that shRNA knockdown of Cav-3 similarly prolonged APD. CONCLUSION We demonstrate that Cav-3 and caveolae regulate cardiac repolarization and arrhythmia risk via the integrated modulation of multiple ionic currents.
Collapse
Affiliation(s)
- Yogananda S Markandeya
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA; National Institute of Mental Health and Neuroscience, Bengaluru, India
| | - Zachery R Gregorich
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Li Feng
- Department of Cardiology, Beijing Anzhen Hospital, Captial Medical University, National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Vignesh Ramchandran
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Thomas O' Hara
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ravi Vaidyanathan
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Catherine Mansfield
- National Heart and Lung Institute, Imperial College London, ICTEM, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Alexis M Keefe
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Carl J Beglinger
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Jabe M Best
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Matthew M Kalscheur
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Martin R Lea
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Timothy A Hacker
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, ICTEM, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Natalia A Trayanova
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lee L Eckhardt
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Jonathan C Makielski
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Ravi C Balijepalli
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Timothy J Kamp
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA.
| |
Collapse
|
9
|
Struckman HL, Moise N, King DR, Soltisz A, Buxton A, Dunlap I, Chen Z, Radwański PB, Weinberg SH, Veeraraghavan R. Unraveling Chamber-specific Differences in Intercalated Disc Ultrastructure and Molecular Organization and Their Impact on Cardiac Conduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528369. [PMID: 36824727 PMCID: PMC9949041 DOI: 10.1101/2023.02.13.528369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
During each heartbeat, the propagation of action potentials through the heart coordinates the contraction of billions of individual cardiomyocytes and is thus, a critical life process. Unsurprisingly, intercalated discs, which are cell-cell contact sites specialized to provide electrical and mechanical coupling between adjacent cardiomyocytes, have been the focus of much investigation. Slowed or disrupted propagation leads to potentially life-threatening arrhythmias in a wide range of pathologies, where intercalated disc remodeling is a common finding. Hence, the importance and urgency of understanding intercalated disc structure and its influence on action potential propagation. Surprisingly, however, conventional modeling approaches cannot predict changes in propagation elicited by perturbations that alter intercalated disc ultrastructure or molecular organization, owing to lack of quantitative structural data at subcellular through nano scales. In order to address this critical gap in knowledge, we sought to quantify intercalated disc structure at these finer spatial scales in the healthy adult mouse heart and relate them to function in a chamber-specific manner as a precursor to understanding the impacts of pathological intercalated disc remodeling. Using super-resolution light microscopy, electron microscopy, and computational image analysis, we provide here the first ever systematic, multiscale quantification of intercalated disc ultrastructure and molecular organization. By incorporating these data into a rule-based model of cardiac tissue with realistic intercalated disc structure, and comparing model predictions of electrical propagation with experimental measures of conduction velocity, we reveal that atrial intercalated discs can support faster conduction than their ventricular counterparts, which is normally masked by inter-chamber differences in myocyte geometry. Further, we identify key ultrastructural and molecular organization features underpinning the ability of atrial intercalated discs to support faster conduction. These data provide the first stepping stone to elucidating chamber-specific impacts of pathological intercalated disc remodeling, as occurs in many arrhythmic diseases.
Collapse
|
10
|
Marchal GA, Remme CA. Subcellular diversity of Nav1.5 in cardiomyocytes: distinct functions, mechanisms and targets. J Physiol 2023; 601:941-960. [PMID: 36469003 DOI: 10.1113/jp283086] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/24/2022] [Indexed: 12/11/2022] Open
Abstract
In cardiomyocytes, the rapid depolarisation of the membrane potential is mediated by the α-subunit of the cardiac voltage-gated Na+ channel (NaV 1.5), encoded by the gene SCN5A. This ion channel allows positively charged Na+ ions to enter the cardiomyocyte, resulting in the fast upstroke of the action potential and is therefore crucial for cardiac excitability and electrical propagation. This essential role is underscored by the fact that dysfunctional NaV 1.5 is associated with high risk for arrhythmias and sudden cardiac death. However, development of therapeutic interventions regulating NaV 1.5 has been limited due to the complexity of NaV 1.5 structure and function and its diverse roles within the cardiomyocyte. In particular, research from the last decade has provided us with increased knowledge on the subcellular distribution of NaV 1.5 as well as the proteins which it interacts with in distinct cardiomyocyte microdomains. We here review these insights, detailing the potential role of NaV 1.5 within subcellular domains as well as its dysfunction in the setting of arrhythmia disorders. We furthermore provide an overview of current knowledge on the pathways involved in (microdomain-specific) trafficking of NaV 1.5, and their potential as novel targets. Unravelling the complexity of NaV 1.5 (dys)function may ultimately facilitate the development of therapeutic strategies aimed at preventing lethal arrhythmias. This is not only of importance for pathophysiological conditions where sodium current is specifically decreased within certain subcellular regions, such as in arrhythmogenic cardiomyopathy and Duchenne muscular dystrophy, but also for other acquired and inherited disorders associated with NaV 1.5.
Collapse
Affiliation(s)
- Gerard A Marchal
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands.,National Institute of Optics, National Research Council (CNR-INO), Sesto Fiorentino, Florence, Italy
| | - Carol Ann Remme
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Eckhardt LL. Arrhythmogenesis and Prolonged Repolarization From Synthetic Opioids: Finally Sorted? J Am Heart Assoc 2022; 11:e025778. [PMID: 35658484 PMCID: PMC9238742 DOI: 10.1161/jaha.122.025778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Lee L Eckhardt
- Department of Medicine University of Wisconsin-Madison Madison WI
| |
Collapse
|
12
|
Coyote-Maestas W, Nedrud D, He Y, Schmidt D. Determinants of trafficking, conduction, and disease within a K + channel revealed through multiparametric deep mutational scanning. eLife 2022; 11:e76903. [PMID: 35639599 PMCID: PMC9273215 DOI: 10.7554/elife.76903] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 05/27/2022] [Indexed: 01/04/2023] Open
Abstract
A long-standing goal in protein science and clinical genetics is to develop quantitative models of sequence, structure, and function relationships to understand how mutations cause disease. Deep mutational scanning (DMS) is a promising strategy to map how amino acids contribute to protein structure and function and to advance clinical variant interpretation. Here, we introduce 7429 single-residue missense mutations into the inward rectifier K+ channel Kir2.1 and determine how this affects folding, assembly, and trafficking, as well as regulation by allosteric ligands and ion conduction. Our data provide high-resolution information on a cotranslationally folded biogenic unit, trafficking and quality control signals, and segregated roles of different structural elements in fold stability and function. We show that Kir2.1 surface trafficking mutants are underrepresented in variant effect databases, which has implications for clinical practice. By comparing fitness scores with expert-reviewed variant effects, we can predict the pathogenicity of 'variants of unknown significance' and disease mechanisms of known pathogenic mutations. Our study in Kir2.1 provides a blueprint for how multiparametric DMS can help us understand the mechanistic basis of genetic disorders and the structure-function relationships of proteins.
Collapse
Affiliation(s)
- Willow Coyote-Maestas
- Department of Biochemistry, Molecular Biology and Biophysics, University of MinnesotaMinneapolisUnited States
| | - David Nedrud
- Department of Biochemistry, Molecular Biology and Biophysics, University of MinnesotaMinneapolisUnited States
| | - Yungui He
- Department of Genetics, Cell Biology and Development, University of MinnesotaMinneapolisUnited States
| | - Daniel Schmidt
- Department of Genetics, Cell Biology and Development, University of MinnesotaMinneapolisUnited States
| |
Collapse
|
13
|
Caveolin-3 and Arrhythmias: Insights into the Molecular Mechanisms. J Clin Med 2022; 11:jcm11061595. [PMID: 35329921 PMCID: PMC8952412 DOI: 10.3390/jcm11061595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023] Open
Abstract
Caveolin-3 is a muscle-specific protein on the membrane of myocytes correlated with a variety of cardiovascular diseases. It is now clear that the caveolin-3 plays a critical role in the cardiovascular system and a significant role in cardiac protective signaling. Mutations in the gene encoding caveolin-3 cause a broad spectrum of clinical phenotypes, ranging from persistent elevations in the serum levels of creatine kinase in asymptomatic humans to cardiomyopathy. The influence of Caveolin-3(CAV-3) mutations on current density parallels the effect on channel trafficking. For example, mutations in the CAV-3 gene promote ventricular arrhythmogenesis in long QT syndrome 9 by a combined decrease in the loss of the inward rectifier current (IK1) and gain of the late sodium current (INa-L). The functional significance of the caveolin-3 has proved that caveolin-3 overexpression or knockdown contributes to the occurrence and development of arrhythmias. Caveolin-3 overexpression could lead to reduced diastolic spontaneous Ca2+ waves, thus leading to the abnormal L-Type calcium channel current-induced ventricular arrhythmias. Moreover, CAV-3 knockdown resulted in a shift to more negative values in the hyperpolarization-activated cyclic nucleotide channel 4 current (IHCN4) activation curve and a significant decrease in IHCN4 whole-cell current density. Recent evidence indicates that caveolin-3 plays a significant role in adipose tissue and is related to obesity development. The role of caveolin-3 in glucose homeostasis has attracted increasing attention. This review highlights the underlining mechanisms of caveolin-3 in arrhythmia. Progress in this field may contribute to novel therapeutic approaches for patients prone to developing arrhythmia.
Collapse
|
14
|
Hager NA, McAtee CK, Lesko MA, O’Donnell AF. Inwardly Rectifying Potassium Channel Kir2.1 and its "Kir-ious" Regulation by Protein Trafficking and Roles in Development and Disease. Front Cell Dev Biol 2022; 9:796136. [PMID: 35223865 PMCID: PMC8864065 DOI: 10.3389/fcell.2021.796136] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Potassium (K+) homeostasis is tightly regulated for optimal cell and organismal health. Failure to control potassium balance results in disease, including cardiac arrythmias and developmental disorders. A family of inwardly rectifying potassium (Kir) channels helps cells maintain K+ levels. Encoded by KCNJ genes, Kir channels are comprised of a tetramer of Kir subunits, each of which contains two-transmembrane domains. The assembled Kir channel generates an ion selectivity filter for K+ at the monomer interface, which allows for K+ transit. Kir channels are found in many cell types and influence K+ homeostasis across the organism, impacting muscle, nerve and immune function. Kir2.1 is one of the best studied family members with well-defined roles in regulating heart rhythm, muscle contraction and bone development. Due to their expansive roles, it is not surprising that Kir mutations lead to disease, including cardiomyopathies, and neurological and metabolic disorders. Kir malfunction is linked to developmental defects, including underdeveloped skeletal systems and cerebellar abnormalities. Mutations in Kir2.1 cause the periodic paralysis, cardiac arrythmia, and developmental deficits associated with Andersen-Tawil Syndrome. Here we review the roles of Kir family member Kir2.1 in maintaining K+ balance with a specific focus on our understanding of Kir2.1 channel trafficking and emerging roles in development and disease. We provide a synopsis of the vital work focused on understanding the trafficking of Kir2.1 and its role in development.
Collapse
Affiliation(s)
| | | | | | - Allyson F. O’Donnell
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
15
|
Brown KA, Anderson C, Reilly L, Sondhi K, Ge Y, Eckhardt LL. Proteomic Analysis of the Functional Inward Rectifier Potassium Channel (Kir) 2.1 Reveals Several Novel Phosphorylation Sites. Biochemistry 2021; 60:3292-3301. [PMID: 34676745 DOI: 10.1021/acs.biochem.1c00555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Membrane proteins represent a large family of proteins that perform vital physiological roles and represent key drug targets. Despite their importance, bioanalytical methods aiming to comprehensively characterize the post-translational modification (PTM) of membrane proteins remain challenging compared to other classes of proteins in part because of their inherent low expression and hydrophobicity. The inward rectifier potassium channel (Kir) 2.1, an integral membrane protein, is critical for the maintenance of the resting membrane potential and phase-3 repolarization of the cardiac action potential in the heart. The importance of this channel to cardiac physiology is highlighted by the recognition of several sudden arrhythmic death syndromes, Andersen-Tawil and short QT syndromes, which are associated with loss or gain of function mutations in Kir2.1, often triggered by changes in the β-adrenergic tone. Therefore, understanding the PTMs of this channel (particularly β-adrenergic tone-driven phosphorylation) is important for arrhythmia prevention. Here, we developed a proteomic method, integrating both top-down (intact protein) and bottom-up (after enzymatic digestion) proteomic analyses, to characterize the PTMs of recombinant wild-type and mutant Kir2.1, successfully mapping five novel sites of phosphorylation and confirming a sixth site. Our study provides a framework for future work to assess the role of PTMs in regulating Kir2.1 functions.
Collapse
Affiliation(s)
- Kyle A Brown
- Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States.,Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Corey Anderson
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Louise Reilly
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Kunal Sondhi
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Lee L Eckhardt
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
16
|
Reilly L, Eckhardt LL. Cardiac potassium inward rectifier Kir2: Review of structure, regulation, pharmacology, and arrhythmogenesis. Heart Rhythm 2021; 18:1423-1434. [PMID: 33857643 PMCID: PMC8328935 DOI: 10.1016/j.hrthm.2021.04.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/23/2021] [Accepted: 04/06/2021] [Indexed: 12/17/2022]
Abstract
Potassium inward rectifier channel Kir2 is an important component of terminal cardiac repolarization and resting membrane stability. This functionality is part of balanced cardiac excitability and is a defining feature of excitable cardiac membranes. “Gain-of-function” or “loss-of-function” mutations in KCNJ2, the gene encoding Kir2.1, cause genetic sudden cardiac death syndromes, and loss of the Kir2 current IK1 is a major contributing factor to arrhythmogenesis in failing human hearts. Here we provide a contemporary review of the functional structure, physiology, and pharmacology of Kir2 channels. Beyond the structure and functional relationships, we will focus on the elements of clinically used drugs that block the channel and the implications for treatment of atrial fibrillation with IK1-blocking agents. We will also review the clinical disease entities associated with KCNJ2 mutations and the growing area of research into associated arrhythmia mechanisms. Lastly, the presence of Kir2 channels has become a tipping point for electrical maturity in induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) and highlights the significance of understanding why Kir2 in iPS-CMs is important to consider for Comprehensive In Vitro Proarrhythmia Assay and drug safety testing.
Collapse
Affiliation(s)
- Louise Reilly
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Lee L Eckhardt
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin.
| |
Collapse
|
17
|
Ivanova AD, Filatova TS, Abramochkin DV, Atkinson A, Dobrzynski H, Kokaeva ZG, Merzlyak EM, Pustovit KB, Kuzmin VS. Attenuation of inward rectifier potassium current contributes to the α1-adrenergic receptor-induced proarrhythmicity in the caval vein myocardium. Acta Physiol (Oxf) 2021; 231:e13597. [PMID: 33306261 DOI: 10.1111/apha.13597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 11/19/2020] [Accepted: 12/07/2020] [Indexed: 12/31/2022]
Abstract
AIM This study is aimed at investigation of electrophysiological effects of α1-adrenoreceptor (α1-AR) stimulation in the rat superior vena cava (SVC) myocardium, which is one of the sources of proarrhythmic activity. METHODS α1-ARs agonists (phenylephrine-PHE or norepinephrine in presence of atenolol-NE + ATL) were applied to SVC and atrial tissue preparations or isolated cardiomyocytes, which were examined using optical mapping, glass microelectrodes or whole-cell patch clamp. α1-ARs distribution was evaluated using immunofluorescence. Kir2.X mRNA and protein level were estimated using RT-PCR and Western blotting. RESULTS PHE or NE + ATL application caused a significant suppression of the conduction velocity (CV) of excitation and inexcitability in SVC, an increase in the duration of electrically evoked action potentials (APs), a decrease in the maximum upstroke velocity (dV/dtmax ) and depolarization of the resting membrane potential (RMP) in SVC to a greater extent than in atria. The effects induced by α1-ARs activation in SVC were attenuated by protein kinase C inhibition (PKC). The whole-cell patch clamp revealed PHE-induced suppression of outward component of IK1 inward rectifier current in isolated SVC, but not atrial myocytes. These effects can be mediated by α1A subtype of α-ARs found in abundance in rat SVC. The basal IK1 level in SVC was much lower than in atria as a result of the weaker expression of Kir2.2 channels. CONCLUSION Therefore, the reduced density of IK1 in rat SVC cardiomyocytes and sensitivity of this current to α1A-AR stimulation via PKC-dependent pathways might lead to proarrhythmic conduction in SVC myocardium by inducing RMP depolarization, AP prolongation, CV and dV/dtmax decrease.
Collapse
Affiliation(s)
- Alexandra D. Ivanova
- Department of Human and Animal Physiology Lomonosov Moscow State University Moscow Russia
| | - Tatiana S. Filatova
- Department of Human and Animal Physiology Lomonosov Moscow State University Moscow Russia
- Department of Physiology Pirogov Russian National Research Medical University Moscow Russia
| | - Denis V. Abramochkin
- Department of Human and Animal Physiology Lomonosov Moscow State University Moscow Russia
- Department of Physiology Pirogov Russian National Research Medical University Moscow Russia
- Laboratory of Cardiac Electrophysiology National Medical Research Center for Cardiology Moscow Russia
| | - Andrew Atkinson
- Faculty of Biology, Medicine and Health University of Manchester Manchester UK
| | - Halina Dobrzynski
- Faculty of Biology, Medicine and Health University of Manchester Manchester UK
- Heart Embryology and Anatomy Research Team Department of Anatomy Jagiellonian University Medical College Cracow Poland
| | - Zarema G. Kokaeva
- Department of Human and Animal Physiology Lomonosov Moscow State University Moscow Russia
| | - Ekaterina M. Merzlyak
- Shemiakin‐Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science Moscow Russia
| | - Ksenia B. Pustovit
- Department of Human and Animal Physiology Lomonosov Moscow State University Moscow Russia
| | - Vladislav S. Kuzmin
- Department of Human and Animal Physiology Lomonosov Moscow State University Moscow Russia
- Department of Physiology Pirogov Russian National Research Medical University Moscow Russia
| |
Collapse
|
18
|
Zaza A, Grandi E. Mechanisms of Cav3-associated arrhythmia: Protein or microdomain dysfunction? Int J Cardiol 2020; 320:97-99. [DOI: 10.1016/j.ijcard.2020.06.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 10/23/2022]
|
19
|
Kim SY, Kim KH, Schilling JM, Leem J, Dhanani M, Head BP, Roth DM, Zemljic-Harpf AE, Patel HH. Protective role of cardiac-specific overexpression of caveolin-3 in cirrhotic cardiomyopathy. Am J Physiol Gastrointest Liver Physiol 2020; 318:G531-G541. [PMID: 31961720 PMCID: PMC7099497 DOI: 10.1152/ajpgi.00346.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cirrhotic cardiomyopathy is a clinical syndrome in patients with liver cirrhosis characterized by blunted cardiac contractile responses to stress and/or heart rate-corrected QT (QTc) interval prolongation. Caveolin-3 (Cav-3) plays a critical role in cardiac protection and is an emerging therapeutic target for heart disease. We investigated the protective role of cardiac-specific overexpression (OE) of Cav-3 in cirrhotic cardiomyopathy. Biliary fibrosis was induced in male Cav-3 OE mice and transgene negative (TGneg) littermates by feeding a diet containing 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC; 0.1%) for 3 wk. Liver pathology and blood chemistries were assessed, and stress echocardiography, telemetry, and isolated heart perfusion studies to assess adrenergic responsiveness were performed. Cav-3 OE mice showed a similar degree of hyperdynamic contractility, pulmonary hypertension, and QTc interval prolongation as TGneg mice after 3 wk of DDC diet. Blunted systolic responses were shown in both DDC-fed Cav-3 OE and TGneg hearts after in vivo isoproterenol challenge. However, QTc interval prolongation after in vivo isoproterenol challenge was significantly less in DDC-fed Cav-3 OE hearts compared with DDC-fed TGneg hearts. In ex vivo perfused hearts, where circulatory factors are absent, isoproterenol challenge showed hearts from DDC-fed Cav-3 OE mice had better cardiac contractility and relaxation compared with DDC-fed TGneg hearts. Although Cav-3 OE in the heart did not prevent cardiac alterations in DDC-induced biliary fibrosis, cardiac expression of Cav-3 reduced QTc interval prolongation after adrenergic stimulation in cirrhosis.NEW & NOTEWORTHY Prevalence of cirrhotic cardiomyopathy is up to 50% in cirrhotic patients, and liver transplantation is the only treatment. However, cirrhotic cardiomyopathy is associated with perioperative morbidity and mortality after liver transplantation; therefore, management of cirrhotic cardiomyopathy is crucial for successful liver transplantation. This study shows cardiac myocyte specific overexpression of caveolin-3 (Cav-3) provides better cardiac contractile responses and less corrected QT prolongation during adrenergic stress in a cirrhotic cardiomyopathy model, suggesting beneficial effects of Cav-3 expression in cirrhotic cardiomyopathy.
Collapse
Affiliation(s)
- So Yeon Kim
- 1Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kang Ho Kim
- 2Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jan M. Schilling
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - Joseph Leem
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - Mehul Dhanani
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - Brian P. Head
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - David M. Roth
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - Alice E. Zemljic-Harpf
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| | - Hemal H. Patel
- 3Veterans Administration San Diego Healthcare System and the Department of Anesthesiology, University of California, San Diego, School of Medicine, San Diego, California
| |
Collapse
|
20
|
Huang Y, Deng Y, Shang L, Yang L, Huang J, Ma J, Liao X, Zhou H, Xian J, Liang G, Huang Q. Effect of type 2 diabetes mellitus caveolin-3 K15N mutation on glycometabolism. Exp Ther Med 2019; 18:2531-2539. [PMID: 31572504 PMCID: PMC6755474 DOI: 10.3892/etm.2019.7840] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 05/23/2019] [Indexed: 01/08/2023] Open
Abstract
Caveolin-3 (CAV3) is a muscle-specific protein present within the muscle cell membrane that affects signaling pathways, including the insulin signaling pathway. A previous assessment of patients with newly developed type 2 diabetes (T2DM) demonstrated that CAV3 gene mutations may lead to changes in protein secondary structure. A severe CAV3 P104L mutation has previously been indicated to influence the phosphorylation of skeletal muscle cells and result in impaired glucose metabolism. In the present study, the effect of CAV3 K15N gene transfection in C2C12 cells was assessed. Transfection with K15N reduced the expression of total CAV3 and AKT2 proteins in the cells, and the translocation of glucose transporter type 4 to the muscle cell membrane, which resulted in decreased glucose uptake and glycogen synthesis in myocytes. In conclusion, these results indicate that the CAV3 K15N mutation may cause insulin-stimulated impaired glucose metabolism in myocytes, which may contribute to the development of T2DM.
Collapse
Affiliation(s)
- Yiyuan Huang
- School of Nursing, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Yufeng Deng
- School of Nursing, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Lina Shang
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Lihui Yang
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Juanjuan Huang
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jing Ma
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xianshan Liao
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Hui Zhou
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jing Xian
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Guining Liang
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qin Huang
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
21
|
Qile M, Ji Y, Houtman MJC, Veldhuis M, Romunde F, Kok B, van der Heyden MAG. Identification of a PEST Sequence in Vertebrate K IR2.1 That Modifies Rectification. Front Physiol 2019; 10:863. [PMID: 31333502 PMCID: PMC6624654 DOI: 10.3389/fphys.2019.00863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/20/2019] [Indexed: 11/13/2022] Open
Abstract
KIR2.1 potassium channels, producing inward rectifier potassium current (IK1), are important for final action potential repolarization and a stable resting membrane potential in excitable cells like cardiomyocytes. Abnormal KIR2.1 function, either decreased or increased, associates with diseases such as Andersen-Tawil syndrome, long and short QT syndromes. KIR2.1 ion channel protein trafficking and subcellular anchoring depends on intrinsic specific short amino acid sequences. We hypothesized that combining an evolutionary based sequence comparison and bioinformatics will identify new functional domains within the C-terminus of the KIR2.1 protein, which function could be determined by mutation analysis. We determined PEST domain signatures, rich in proline (P), glutamic acid (E), serine (S), and threonine (T), within KIR2.1 sequences using the “epestfind” webtool. WT and ΔPEST KIR2.1 channels were expressed in HEK293T and COS-7 cells. Patch-clamp electrophysiology measurements were performed in the inside-out mode on excised membrane patches and the whole cell mode using AxonPatch 200B amplifiers. KIR2.1 protein expression levels were determined by western blot analysis. Immunofluorescence microscopy was used to determine KIR2.1 subcellular localization. An evolutionary conserved PEST domain was identified in the C-terminus of the KIR2.1 channel protein displaying positive PEST scores in vertebrates ranging from fish to human. No similar PEST domain was detected in KIR2.2, KIR2.3, and KIR2.6 proteins. Deletion of the PEST domain in California kingsnake and human KIR2.1 proteins (ΔPEST), did not affect plasma membrane localization. Co-expression of WT and ΔPEST KIR2.1 proteins resulted in heterotetrameric channel formation. Deletion of the PEST domain did not increase protein stability in cycloheximide assays [T½ from 2.64 h (WT) to 1.67 h (ΔPEST), n.s.]. WT and ΔPEST channels, either from human or snake, produced typical IK1, however, human ΔPEST channels displayed stronger intrinsic rectification. The current observations suggest that the PEST sequence of KIR2.1 is not associated with rapid protein degradation, and has a role in the rectification behavior of IK1 channels.
Collapse
Affiliation(s)
- Muge Qile
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Yuan Ji
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marien J C Houtman
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marlieke Veldhuis
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Fee Romunde
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Bart Kok
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marcel A G van der Heyden
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
22
|
Chasing Uptake: Super-Resolution Microscopy in Endocytosis and Phagocytosis. Trends Cell Biol 2019; 29:727-739. [PMID: 31227311 DOI: 10.1016/j.tcb.2019.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/14/2019] [Accepted: 05/23/2019] [Indexed: 11/21/2022]
Abstract
Since their invention about two decades ago, super-resolution microscopes have become a method of choice in cell biology. Owing to a spatial resolution below 50 nm, smaller than the size of most organelles, and an order of magnitude better than the diffraction limit of conventional light microscopes, super-resolution microscopy is a powerful technique for resolving intracellular trafficking. In this review we discuss discoveries in endocytosis and phagocytosis that have been made possible by super-resolution microscopy - from uptake at the plasma membrane, endocytic coat formation, and cytoskeletal rearrangements to endosomal maturation. The detailed visualization of the diverse molecular assemblies that mediate endocytic uptake will provide a better understanding of how cells ingest extracellular material.
Collapse
|
23
|
Nanoscale reorganization of sarcoplasmic reticulum in pressure-overload cardiac hypertrophy visualized by dSTORM. Sci Rep 2019; 9:7867. [PMID: 31133706 PMCID: PMC6536555 DOI: 10.1038/s41598-019-44331-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023] Open
Abstract
Pathological cardiac hypertrophy is a debilitating condition characterized by deleterious thickening of the myocardium, dysregulated Ca2+ signaling within cardiomyocytes, and contractile dysfunction. Importantly, the nanoscale organization, localization, and patterns of expression of critical Ca2+ handling regulators including dihydropyridine receptor (DHPR), ryanodine receptor 2 (RyR2), phospholamban (PLN), and sarco/endoplasmic reticulum Ca2+-ATPase 2A (SERCA2A) remain poorly understood, especially during pathological hypertrophy disease progression. In the current study, we induced cardiac pathological hypertrophy via transverse aortic constriction (TAC) on 8-week-old CD1 mice, followed by isolation of cardiac ventricular myocytes. dSTORM super-resolution imaging was then used to visualize proteins at nanoscale resolution at two time points and we quantified changes in protein cluster properties using Voronoi tessellation and 2D Fast Fourier Transform analyses. We showed a decrease in the density of DHPR and RyR2 clusters with pressure-overload cardiac hypertrophy and an increase in the density of SERCA2A protein clusters. PLN protein clusters decreased in density in 2-week TAC but returned to sham levels by 4-week TAC. Furthermore, 2D-FFT analysis revealed changes in molecular organization during pathological hypertrophy, with DHPR and RyR2 becoming dispersed while both SERCA2A and PLN sequestered into dense clusters. Our work reveals molecular adaptations that occur in critical SR proteins at a single molecule during pressure overload-induced cardiomyopathy. Nanoscale alterations in protein localization and patterns of expression of crucial SR proteins within the cardiomyocyte provided insights into the pathogenesis of cardiac hypertrophy, and specific evidence that cardiomyocytes undergo significant structural remodeling during the progression of pathological hypertrophy.
Collapse
|
24
|
Tyan L, Foell JD, Vincent KP, Woon MT, Mesquitta WT, Lang D, Best JM, Ackerman MJ, McCulloch AD, Glukhov AV, Balijepalli RC, Kamp TJ. Long QT syndrome caveolin-3 mutations differentially modulate K v 4 and Ca v 1.2 channels to contribute to action potential prolongation. J Physiol 2019; 597:1531-1551. [PMID: 30588629 DOI: 10.1113/jp276014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 12/14/2018] [Indexed: 01/09/2023] Open
Abstract
KEY POINTS Mutations in the caveolae scaffolding protein, caveolin-3 (Cav3), have been linked to the long QT type 9 inherited arrhythmia syndrome (LQT9) and the cause of underlying action potential duration prolongation is incompletely understood. In the present study, we show that LQT9 Cav3 mutations, F97C and S141R, cause mutation-specific gain of function effects on Cav 1.2-encoded L-type Ca2+ channels responsible for ICa,L and also cause loss of function effects on heterologously expressed Kv 4.2 and Kv 4.3 channels responsible for Ito . A computational model of the human ventricular myocyte action potential suggests that the major ionic current change causing action potential duration prolongation in the presence of Cav3-F97C is the slowly inactivating ICa,L but, for Cav3-S141R, both increased ICa,L and increased late Na+ current contribute equally to action potential duration prolongation. Overall, the LQT9 Cav3-F97C and Cav3-S141R mutations differentially impact multiple ionic currents, highlighting the complexity of Cav3 regulation of cardiac excitability and suggesting mutation-specific therapeutic approaches. ABSTRACT Mutations in the CAV3 gene encoding caveolin-3 (Cav3), a scaffolding protein integral to caveolae in cardiomyocytes, have been associated with the congenital long-QT syndrome (LQT9). Initial studies demonstrated that LQT9-associated Cav3 mutations, F97C and S141R, increase late sodium current as a potential mechanism to prolong action potential duration (APD) and cause LQT9. Whether these Cav3 LQT9 mutations impact other caveolae related ion channels remains unknown. We used the whole-cell, patch clamp technique to characterize the effect of Cav3-F97C and Cav3-S141R mutations on heterologously expressed Cav 1.2+Cav β2cN4 channels, as well as Kv 4.2 and Kv 4.3 channels, in HEK 293 cells. Expression of Cav3-S141R increased ICa,L density without changes in gating properties, whereas expression of Cav3-F97C reduced Ca2+ -dependent inactivation of ICa,L without changing current density. The Cav3-F97C mutation reduced current density and altered the kinetics of IKv4.2 and IKv4.3 and also slowed recovery from inactivation. Cav3-S141R decreased current density and also slowed activation kinetics and recovery from inactivation of IKv4.2 but had no effect on IKv4.3 . Using the O'Hara-Rudy computational model of the human ventricular myocyte action potential, the Cav3 mutation-induced changes in Ito are predicted to have negligible effect on APD, whereas blunted Ca2+ -dependent inactivation of ICa,L by Cav3-F97C is predicted to be primarily responsible for APD prolongation, although increased ICa,L and late INa by Cav3-S141R contribute equally to APD prolongation. Thus, LQT9 Cav3-associated mutations, F97C and S141R, produce mutation-specific changes in multiple ionic currents leading to different primary causes of APD prolongation, which suggests the use of mutation-specific therapeutic approaches in the future.
Collapse
Affiliation(s)
- Leonid Tyan
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, 1111, Highland Ave, Madison, WI, USA
| | - Jason D Foell
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, 1111, Highland Ave, Madison, WI, USA
| | - Kevin P Vincent
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, USA
| | - Marites T Woon
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, 1111, Highland Ave, Madison, WI, USA
| | - Walatta T Mesquitta
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, 1111, Highland Ave, Madison, WI, USA
| | - Di Lang
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, 1111, Highland Ave, Madison, WI, USA
| | - Jabe M Best
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, 1111, Highland Ave, Madison, WI, USA
| | - Michael J Ackerman
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine and Molecular Pharmacology & Experimental Therapeutics, Divisions of Heart Rhythm Services and Pediatric Cardiology, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, USA.,Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, USA
| | - Alexey V Glukhov
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, 1111, Highland Ave, Madison, WI, USA
| | - Ravi C Balijepalli
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, 1111, Highland Ave, Madison, WI, USA
| | - Timothy J Kamp
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, 1111, Highland Ave, Madison, WI, USA
| |
Collapse
|
25
|
Vaidyanathan R, Reilly L, Eckhardt LL. Caveolin-3 Microdomain: Arrhythmia Implications for Potassium Inward Rectifier and Cardiac Sodium Channel. Front Physiol 2018; 9:1548. [PMID: 30473666 PMCID: PMC6238080 DOI: 10.3389/fphys.2018.01548] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/16/2018] [Indexed: 11/13/2022] Open
Abstract
In human cardiac ventricular myocytes, caveolin-3 functions as a scaffolding and regulatory protein for signaling molecules and compartmentalizes ion channels. Our lab has recently explored this sub-cellular microdomain and found that potassium inward rectifier Kir2.x is found in association with caveolin-3. The three cardiac Kir2.x isoforms (Kir2.1, Kir2.2, and Kir2.3) are the molecular correlates of IK1 in the heart, of which Kir2.1 is the dominant isoform in the ventricle. Kir2.1 channels assemble with Kir2.2 and Kir2.3 forming hetero-tetramers that modulate IK1. IK1 sets the resting membrane potential and assists with terminal phase 3 ventricular repolarization. In our studies using native human ventricular tissue, Kir2.x co-localizes with caveolin-3 and significance of the association between Kir2.x and caveolin-3 is emphasized in relation to mutations in the gene which encodes caveolin-3, CAV3, associated with Long QT Syndrome 9 (LQT9). LQT9-associated CAV3 mutations cause decreased current density in Kir2.1 and Kir2.2 as homomeric and heteromeric channels, which affects repolarization and membrane potential stability. A portion of Kir2.1 cardiac localization parallels that of the cardiac sodium channel (Nav1.5). This may have implications for Long QT9 in which CAV3 mutations cause an increase in the late current of Nav1.5 (INa-L) via nNOS mediated nitrosylation of Nav1.5. In iPS-CMs, expression of LQT9 CAV3 mutations resulted in action potential duration (APD) prolongation and early-after depolarizations (EADs), supporting the arrhythmogenicity of LQT9. To evaluate the combined effect of the CAV3 mutants on INa-L and IK1, we studied both ventricular and Purkinje myocyte mathematical modeling. Interestingly, mathematical ventricular myocytes, similar to iPS-CMs, demonstrated EADs but no sustained arrhythmia. In contrast, Purkinje modeling demonstrated delayed-after depolarizations (DADs) driven mechanism for sustained arrhythmia, dependent on the combined loss of IK1 and gain of INa-L. This finding changes the overall assumed arrhythmia phenotype for LQT9. In future studies, we are exploring caveolar micro-domain disruption in heart failure and how this effects Kir2.x and Nav1.5. Here we review the caveolae cardiac microdomain of Kir2.x and Nav1.5 and explore some of the downstream effects of caveolin-3 and caveolae disruption in specific clinical scenarios.
Collapse
Affiliation(s)
- Ravi Vaidyanathan
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Louise Reilly
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Lee L Eckhardt
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
26
|
Gong X, Han X, Lu X, Chen J, Huang J, Kelly TN, Chen CS, He J, Gu D, Chen S. Association of Kir genes with blood pressure responses to dietary sodium intervention: the GenSalt study. Hypertens Res 2018; 41:1045-1053. [PMID: 30323262 DOI: 10.1038/s41440-018-0113-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/19/2018] [Accepted: 05/12/2018] [Indexed: 12/19/2022]
Abstract
Blood pressure (BP) responses to dietary sodium intervention vary among individuals. The inwardly rectifying potassium channel (Kir) is a potassium-selective ion channel that allows potassium ions to move more easily into rather than out of the cell. We aimed to investigate the associations of Kir genes with BP responses to dietary sodium intervention. A 7-day low-sodium intervention followed by a 7-day high-sodium intervention was conducted among 1906 participants. BP measurements were obtained at baseline and during each dietary intervention. Both single-marker and gene-based analyses were performed to explore the associations between Kir gene variants and BP responses to dietary sodium interventions. The genetic risk score (GRS) was used to assess the cumulative effect of the variants on the BP response to the sodium interventions. During the low-sodium intervention, markers rs858009, rs2835904, and rs860795 in KCNJ6 were significantly associated with the systolic BP (SBP) response (P = 8.82 × 10-6, 3.32 × 10-6, and 2.39 × 10-4, respectively), whereas rs858009 and rs2835904 were significantly correlated with the mean arterial pressure (MAP) response (P = 1.64 × 10-4 and 2.72 × 10-4, respectively). Marker rs2836023 showed a significant association with the SBP response (P = 5.72 × 10-5) to the high-sodium intervention. The GRS stratified by quartile grouping or as a continuous variable was associated with the BP response to the sodium interventions. Gene-based analyses consistently revealed that KCNJ6 was significantly associated with the BP response to the sodium interventions. These findings suggest that KCNJ6 may contribute to variation of BP responses to dietary sodium interventions. Future studies are warranted to confirm these findings and to identify functional variants for salt sensitivity.
Collapse
Affiliation(s)
- Xinyuan Gong
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xikun Han
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangfeng Lu
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Jianfeng Huang
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Chung-Shiuan Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Dongfeng Gu
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shufeng Chen
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
27
|
Taneja TK, Ma D, Kim BY, Welling PA. Golgin-97 Targets Ectopically Expressed Inward Rectifying Potassium Channel, Kir2.1, to the trans-Golgi Network in COS-7 Cells. Front Physiol 2018; 9:1070. [PMID: 30123141 PMCID: PMC6085455 DOI: 10.3389/fphys.2018.01070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/17/2018] [Indexed: 12/27/2022] Open
Abstract
The inward rectifying potassium channel, Kir2.1, is selected as cargo at the trans-Golgi network (TGN) for export to the cell surface through a unique signal-dependent interaction with the AP1 clathrin-adaptor, but it is unknown how the channel is targeted at earlier stages in the secretory pathway for traffic to the TGN. Here we explore a mechanism. A systematic screen of Golgi tethers identified Golgin-97 as a Kir2.1 binding partner. In vitro protein-interaction studies revealed the interaction is direct, occurring between the GRIP domain of Golgin-97 and the cytoplasmic domain of Kir2.1. Imaging and interaction studies in COS-7 cells suggest that Golgi-97 binds to the channel en route through the Golgi. RNA interference-mediated knockdown of Golgin-97 prevented exit of Kir2.1 from the Golgi. These observations identify Golgin-97 as a Kir2.1 binding partner that is required for targeting the channel to the TGN. Based on our studies in COS-7 cells, we propose Golgi-97 facilitates formation of AP1-dependent export carriers for Kir2.1 by coupling anterograde delivery of Kir2.1 with retrograde recycling of AP-1 containing endosomes to the TGN.
Collapse
Affiliation(s)
- Tarvinder K Taneja
- Department of Physiology, Maryland Center for Kidney Discovery, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Donghui Ma
- Department of Physiology, Maryland Center for Kidney Discovery, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Bo Y Kim
- Department of Physiology, Maryland Center for Kidney Discovery, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Paul A Welling
- Department of Physiology, Maryland Center for Kidney Discovery, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| |
Collapse
|