1
|
Biswas R, López-Serrano AL, Purohit A, Ramirez-Navarro A, Huang HL, Grandinetti G, Cheng X, Heissler SM, Deschênes I, Chinthalapudi K. Structural basis of human Na v1.5 gating mechanisms. Proc Natl Acad Sci U S A 2025; 122:e2416181122. [PMID: 40366698 PMCID: PMC12107192 DOI: 10.1073/pnas.2416181122] [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: 08/09/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Voltage-gated Nav1.5 channels are central to the generation and propagation of cardiac action potentials. Aberrations in their function are associated with a wide spectrum of cardiac diseases including arrhythmias and heart failure. Despite decades of progress in Nav1.5 biology, the lack of structural insights into intracellular regions has hampered our understanding of its gating mechanisms. Here, we present two cryo-EM structures of human Nav1.5 in open states, revealing sequential conformational changes in gating charges of the voltage-sensing domains (VSDs) and several intracellular regions. Despite the channel being in the open state, these structures show repositioning, but no dislodging of the IFM motif in the receptor site. Molecular dynamics analyses show our structures with CTD conduct Na+ ions. Notably, our structural findings highlight a dynamic C-terminal domain (CTD) and III-IV linker interaction, which regulates the conformation of VSDs and pore opening. Electrophysiological studies confirm that disrupting this interaction alters fast inactivation of Nav1.5. Together, our structure-function studies establish a foundation for understanding the gating mechanisms of Nav1.5 and the mechanisms underlying CTD-related channelopathies.
Collapse
Affiliation(s)
- Rupam Biswas
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Ana Laura López-Serrano
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH43210
| | - Apoorva Purohit
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH43210
| | - Angelina Ramirez-Navarro
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH43210
| | - Hsiang-Ling Huang
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Giovanna Grandinetti
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Center for Electron Microscopy and Analysis, College of Engineering, The Ohio State University, Columbus, OH43210
| | - Xiaolin Cheng
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH43210
- Translational Data Analytics Institute at The Ohio State University, Columbus, OH43210
| | - Sarah M. Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Isabelle Deschênes
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH43210
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| |
Collapse
|
2
|
Yoshinaga D, Craven I, Feng R, Prondzynski M, Shani K, Tharani Y, Mayourian J, Joseph M, Walker D, Bortolin RH, Carreon CK, Boss B, Upton S, Parker KK, Pu WT, Bezzerides VJ. Dysregulation of N-terminal acetylation causes cardiac arrhythmia and cardiomyopathy. Nat Commun 2025; 16:3604. [PMID: 40234403 PMCID: PMC12000442 DOI: 10.1038/s41467-025-58539-2] [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: 08/28/2024] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
N-terminal acetyltransferases including NAA10 catalyze N-terminal acetylation, an evolutionarily conserved co- and post-translational modification. However, little is known about the role of N-terminal acetylation in cardiac homeostasis. To gain insight into cardiac-dependent NAA10 function, we studied a previously unidentified NAA10 variant p.(Arg4Ser) segregating with QT-prolongation, cardiomyopathy, and developmental delay in a large kindred. Here, we show that the NAA10R4S variant reduced enzymatic activity, decreased NAA10-NAA15 complex formation, and destabilized the enzymatic complex N-terminal acetyltransferase A. In NAA10R4S/Y-induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs), dysregulation of the late sodium and slow delayed rectifier potassium currents caused severe repolarization abnormalities, consistent with clinical QT prolongation. Engineered heart tissues generated from NAA10R4S/Y-iPSC-CMs had significantly decreased contractile force and sarcomeric disorganization, consistent with the pedigree's cardiomyopathic phenotype. Proteomic studies revealed dysregulation of metabolic pathways and cardiac structural proteins. We identified small molecule and genetic therapies that normalized the phenotype of NAA10R4S/Y-iPSC-CMs. Our study defines the roles of N-terminal acetylation in cardiac regulation and delineates mechanisms underlying QT prolongation, arrhythmia, and cardiomyopathy caused by NAA10 dysfunction.
Collapse
Affiliation(s)
- Daisuke Yoshinaga
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabel Craven
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rui Feng
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maksymilian Prondzynski
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kevin Shani
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Allston, MA, USA
| | - Yashasvi Tharani
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua Mayourian
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Milosh Joseph
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - David Walker
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raul H Bortolin
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Bridget Boss
- Department of Pediatric Cardiology, Dartmouth Hitchcock Medical Center, Manchester, NH, USA
| | - Sheila Upton
- Department of Medical Genetics, Dartmouth Hitchcock Medical Center, Manchester, NH, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Allston, MA, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vassilios J Bezzerides
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Pediatric Cardiology, Dartmouth Hitchcock Medical Center, Manchester, NH, USA.
| |
Collapse
|
3
|
Wang Q, Deng Y, Fan LL, Dong Y, Zhang AQ, Liu YX. Novel heterozygous mutation of CACNA2D1 gene in a Chinese family with arrhythmia. BMC Cardiovasc Disord 2024; 24:527. [PMID: 39354346 PMCID: PMC11443683 DOI: 10.1186/s12872-024-04204-3] [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: 05/22/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Primary electrical disorders (PEDs) are a group of cardiac rhythm abnormalities that occur in the absence of detectable structural heart disease and are a significant cause of sudden cardiac death (SCD). The initiation of cardiac muscle contraction and relaxation is orchestrated by the action potential (AP), generated through ionic changes across the membrane. Mutations in the AP-related gene CACNA2D1 have been identified as a causative factor for PED. METHODS We recruited a Chinese family with a history of arrhythmia. The proband has experienced palpitations and chest tightness for over 40 years, with symptoms worsening over the past year. Whole exome sequencing (WES) was used to determine the genetic etiologies in this family. RESULTS A novel heterozygous missense mutation (NM_000722.3: c.1685G > C;p.G562A) of CACNA2D1 gene was detected. Genotyping of the proband's parents indicated that the arrhythmia phenotype in the proband was caused by a de novo mutation. CONCLUSIONS WES was utilized to explore the genetic etiology in a family with arrhythmia, leading to the identification of a novel mutation in the CACNA2D1 gene. This study not only expands the mutation spectrum of the CACNA2D1 gene but also contributes to genetic counseling and clinical diagnosis for this family.
Collapse
Affiliation(s)
- Qian Wang
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, China
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
- Department of Operation Center, The Second Xiangya Hospital of Central South University, Changsha, China
- School of Life Sciences, Central South University, Changsha, China
| | - Yong Deng
- Department of Operation Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liang-Liang Fan
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, China
- School of Life Sciences, Central South University, Changsha, China
| | - Yi Dong
- School of Life Sciences, Central South University, Changsha, China
| | - Ai-Qian Zhang
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, China.
| | - Yu-Xing Liu
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China.
- Department of Operation Center, The Second Xiangya Hospital of Central South University, Changsha, China.
- School of Life Sciences, Central South University, Changsha, China.
| |
Collapse
|
4
|
Jin X, Huang J, Wang H, Wang K, Yan N. A versatile residue numbering scheme for Na v and Ca v channels. Cell Chem Biol 2024; 31:1394-1404. [PMID: 39151406 DOI: 10.1016/j.chembiol.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 08/19/2024]
Abstract
Voltage-gated sodium (Nav) and calcium (Cav) channels are responsible for the initiation of electrical signals. They have long been targeted for the treatment of various diseases. The mounting number of cryoelectron microscopy (cryo-EM) structures for diverse subtypes of Nav and Cav channels from multiple organisms necessitates a generic residue numbering system to establish the structure-function relationship and to aid rational drug design or optimization. Here we suggest a structure-based residue numbering scheme, centering around the most conserved residues on each of the functional segments. We elaborate the generic numbers through illustrative examples, focusing on representative drug-binding sites of eukaryotic Nav and Cav channels. We also extend the numbering scheme to compare common disease mutations among different Nav subtypes. Application of the generic residue numbering scheme affords immediate insights into hotspots for pathogenic mutations and critical loci for drug binding and will facilitate drug discovery targeting Nav and Cav channels.
Collapse
Affiliation(s)
- Xueqin Jin
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| | - Huan Wang
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kan Wang
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation, Guangming District, Shenzhen, Guangdong Province 518107, China; Shenzhen Bay Laboratory, Guangming District, Shenzhen, Guangdong Province 518132, China.
| |
Collapse
|
5
|
Goodchild SJ, Ahern CA. Conformational photo-trapping in Na V1.5: Inferring local motions at the "inactivation gate". Biophys J 2024; 123:2167-2175. [PMID: 38664963 PMCID: PMC11309974 DOI: 10.1016/j.bpj.2024.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/22/2024] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
Rapid and effectual inactivation in voltage-gated sodium channels is required for canonical action-potential firing. This "fast" inactivation arises from swift and reversible protein conformational changes that utilize transmembrane segments and the cytoplasmic linker between channel domains III and IV. Until recently, fast inactivation had been accepted to rely on a "ball-and-chain" mechanism whereby a hydrophobic triplet of DIII-IV amino acids (IFM) impairs conductance by binding to a site in central pore of the channel made available by channel opening. New structures of sodium channels have upended this model. Specifically, cryo-electron microscopic structures of eukaryotic sodium channels depict a peripheral binding site for the IFM motif, outside of the pore, opening the possibility of a yet unidentified allosteric mechanism of fast-inactivation gating. We set out to study fast inactivation by photo-trapping human sodium channels in various functional states under voltage control. This was achieved by genetically encoding the crosslinking unnatural amino acid benzophenone phenylalanine at various sites within the DIII-IV linker in the cardiac sodium channel NaV1.5. These data show dynamic state- and positional-dependent trapping of the transient conformations associated with fast inactivation, each yielding different phenotypes and rates of trapping. These data reveal distinct conformational changes that underlie fast inactivation and point to a dynamic environment around the IFM locus.
Collapse
Affiliation(s)
- Samuel J Goodchild
- Department of Anesthesiology, Pharmacology and Therapeutics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher A Ahern
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
6
|
de Lima Conceição MR, Teixeira-Fonseca JL, Marques LP, Souza DS, da Silva Alcântara F, Orts DJB, Roman-Campos D. Extracellular acidification reveals the antiarrhythmic properties of amiodarone related to late sodium current-induced atrial arrhythmia. Pharmacol Rep 2024; 76:585-599. [PMID: 38619735 DOI: 10.1007/s43440-024-00597-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Amiodarone (AMIO) is an antiarrhythmic drug with the pKa in the physiological range. Here, we explored how mild extracellular pH (pHe) changes shape the interaction of AMIO with atrial tissue and impact its pharmacological properties in the classical model of sea anemone sodium channel neurotoxin type 2 (ATX) induced late sodium current (INa-Late) and arrhythmias. METHOD Isolated atrial cardiomyocytes from male Wistar rats and human embryonic kidney cells expressing SCN5A Na+ channels were used for patch-clamp experiments. Isolated right atria (RA) and left atria (LA) tissue were used for bath organ experiments. RESULTS A more acidophilic pHe caused negative inotropic effects on isolated RA and LA atrial tissue, without modification of the pharmacological properties of AMIO. A pHe of 7.0 changed the sodium current (INa) related components of the action potential (AP), which was enhanced in the presence of AMIO. ATXinduced arrhythmias in isolated RA and LA. Also, ATX prolonged the AP duration and enhanced repolarization dispersion in isolated cardiomyocytes in both pHe 7.4 and pHe 7.0. Pre-incubation of the isolated RA and LA and isolated atrial cardiomyocytes with AMIO prevented arrhythmias induced by ATX only at a pHe of 7.0. Moreover, AMIO was able to block INa-Late induced by ATX only at a pHe of 7.0. CONCLUSION The pharmacological properties of AMIO concerning healthy rat atrial tissue are not dependent on pHe. However, the prevention of arrhythmias induced by INa-Late is pHe-dependent. The development of drugs analogous to AMIO with charge stabilization may help to create more effective drugs to treat arrhythmias related to the INa-Late.
Collapse
Affiliation(s)
- Michael Ramon de Lima Conceição
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil
| | - Jorge Lucas Teixeira-Fonseca
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil
| | - Leisiane Pereira Marques
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil
| | - Diego Santos Souza
- Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Fabiana da Silva Alcântara
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil
| | - Diego Jose Belato Orts
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil
| | - Danilo Roman-Campos
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil.
| |
Collapse
|
7
|
Rinné S, Kiper AK, Jacob R, Ortiz-Bonnin B, Schindler RF, Fischer S, Komadowski M, De Martino E, Schäfer MKH, Cornelius T, Fabritz L, Helker CS, Brand T, Decher N. Popeye domain containing proteins modulate the voltage-gated cardiac sodium channel Nav1.5. iScience 2024; 27:109696. [PMID: 38689644 PMCID: PMC11059135 DOI: 10.1016/j.isci.2024.109696] [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: 09/25/2023] [Revised: 12/15/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
Popeye domain containing (POPDC) proteins are predominantly expressed in the heart and skeletal muscle, modulating the K2P potassium channel TREK-1 in a cAMP-dependent manner. POPDC1 and POPDC2 variants cause cardiac conduction disorders with or without muscular dystrophy. Searching for POPDC2-modulated ion channels using a functional co-expression screen in Xenopus oocytes, we found POPDC proteins to modulate the cardiac sodium channel Nav1.5. POPDC proteins downregulate Nav1.5 currents in a cAMP-dependent manner by reducing the surface expression of the channel. POPDC2 and Nav1.5 are both expressed in different regions of the murine heart and consistently POPDC2 co-immunoprecipitates with Nav1.5 from native cardiac tissue. Strikingly, the knock-down of popdc2 in embryonic zebrafish caused an increased upstroke velocity and overshoot of cardiac action potentials. The POPDC modulation of Nav1.5 provides a new mechanism to regulate cardiac sodium channel densities under sympathetic stimulation, which is likely to have a functional impact on cardiac physiology and inherited arrhythmias.
Collapse
Affiliation(s)
- Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 30537 Marburg, Germany
| | - Aytug K. Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 30537 Marburg, Germany
| | - Ralf Jacob
- Institute of Cytobiology, Center for Synthetic Microbiology, Philipps-University of Marburg, 35043 Marburg, Germany
| | - Beatriz Ortiz-Bonnin
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 30537 Marburg, Germany
| | - Roland F.R. Schindler
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Sabine Fischer
- Faculty of Biology, Cell Signaling and Dynamics, Philipps-University Marburg, 35043 Marburg, Germany
| | - Marlene Komadowski
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 30537 Marburg, Germany
| | - Emilia De Martino
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 30537 Marburg, Germany
| | - Martin K.-H. Schäfer
- Institute of Anatomy and Cell Biology, Philipps-University of Marburg, 35037 Marburg, Germany
| | - Tamina Cornelius
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 30537 Marburg, Germany
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences University of Birmingham, Birmingham B15 2TT, UK
- University Center of Cardiovascular Sciences & Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg Eppendorf, 20251 Hamburg and DZHK Hamburg/Kiel/Lübeck, Germany
| | - Christian S.M. Helker
- Faculty of Biology, Cell Signaling and Dynamics, Philipps-University Marburg, 35043 Marburg, Germany
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 30537 Marburg, Germany
| |
Collapse
|
8
|
Zaytseva AK, Kulichik OE, Kostareva AA, Zhorov BS. Biophysical mechanisms of myocardium sodium channelopathies. Pflugers Arch 2024; 476:735-753. [PMID: 38424322 DOI: 10.1007/s00424-024-02930-3] [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: 09/28/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Genetic variants of gene SCN5A encoding the alpha-subunit of cardiac voltage-gated sodium channel Nav1.5 are associated with various diseases, including long QT syndrome (LQT3), Brugada syndrome (BrS1), and progressive cardiac conduction disease (PCCD). In the last decades, the great progress in understanding molecular and biophysical mechanisms of these diseases has been achieved. The LQT3 syndrome is associated with gain-of-function of sodium channels Nav1.5 due to impaired inactivation, enhanced activation, accelerated recovery from inactivation or the late current appearance. In contrast, BrS1 and PCCD are associated with the Nav1.5 loss-of-function, which in electrophysiological experiments can be manifested as reduced current density, enhanced fast or slow inactivation, impaired activation, or decelerated recovery from inactivation. Genetic variants associated with congenital arrhythmias can also disturb interactions of the Nav1.5 channel with different proteins or drugs and cause unexpected reactions to drug administration. Furthermore, mutations can affect post-translational modifications of the channels and their sensitivity to pH and temperature. Here we briefly review the current knowledge on biophysical mechanisms of LQT3, BrS1 and PCCD. We focus on limitations of studies that use heterologous expression systems and induced pluripotent stem cells (iPSC) derived cardiac myocytes and summarize our understanding of genotype-phenotype relations of SCN5A mutations.
Collapse
Affiliation(s)
- Anastasia K Zaytseva
- Almazov National Medical Research Centre, St. Petersburg, Russia.
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia.
| | - Olga E Kulichik
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | | | - Boris S Zhorov
- Almazov National Medical Research Centre, St. Petersburg, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- McMaster University, Hamilton, Canada
| |
Collapse
|
9
|
Biswas R, López-Serrano A, Huang HL, Ramirez-Navarro A, Grandinetti G, Heissler S, Deschênes I, Chinthalapudi K. Structural basis of human Na v1.5 gating mechanisms. RESEARCH SQUARE 2024:rs.3.rs-3985999. [PMID: 38659812 PMCID: PMC11042394 DOI: 10.21203/rs.3.rs-3985999/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Voltage-gated Nav1.5 channels are central to the generation and propagation of cardiac action potentials1. Aberrations in their function are associated with a wide spectrum of cardiac diseases including arrhythmias and heart failure2-5. Despite decades of progress in Nav1.5 biology6-8, the lack of structural insights into intracellular regions has hampered our understanding of its gating mechanisms. Here we present three cryo-EM structures of human Nav1.5 in previously unanticipated open states, revealing sequential conformational changes in gating charges of the voltage-sensing domains (VSDs) and several intracellular regions. Despite the channel being in the open state, these structures show the IFM motif repositioned in the receptor site but not dislodged. In particular, our structural findings highlight a dynamic C-terminal domain (CTD) and III-IV linker interaction, which regulates the conformation of VSDs and pore opening. Electrophysiological studies confirm that disrupting this interaction results in the fast inactivation of Nav1.5. Together, our structure-function studies establish a foundation for understanding the gating mechanisms of Nav1.5 and the mechanisms underlying CTD-related channelopathies.
Collapse
|
10
|
Badura K, Buławska D, Dąbek B, Witkowska A, Lisińska W, Radzioch E, Skwira S, Młynarska E, Rysz J, Franczyk B. Primary Electrical Heart Disease-Principles of Pathophysiology and Genetics. Int J Mol Sci 2024; 25:1826. [PMID: 38339103 PMCID: PMC10855675 DOI: 10.3390/ijms25031826] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/27/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Primary electrical heart diseases, often considered channelopathies, are inherited genetic abnormalities of cardiomyocyte electrical behavior carrying the risk of malignant arrhythmias leading to sudden cardiac death (SCD). Approximately 54% of sudden, unexpected deaths in individuals under the age of 35 do not exhibit signs of structural heart disease during autopsy, suggesting the potential significance of channelopathies in this group of age. Channelopathies constitute a highly heterogenous group comprising various diseases such as long QT syndrome (LQTS), short QT syndrome (SQTS), idiopathic ventricular fibrillation (IVF), Brugada syndrome (BrS), catecholaminergic polymorphic ventricular tachycardia (CPVT), and early repolarization syndromes (ERS). Although new advances in the diagnostic process of channelopathies have been made, the link between a disease and sudden cardiac death remains not fully explained. Evolving data in electrophysiology and genetic testing suggest previously described diseases as complex with multiple underlying genes and a high variety of factors associated with SCD in channelopathies. This review summarizes available, well-established information about channelopathy pathogenesis, genetic basics, and molecular aspects relative to principles of the pathophysiology of arrhythmia. In addition, general information about diagnostic approaches and management is presented. Analyzing principles of channelopathies and their underlying causes improves the understanding of genetic and molecular basics that may assist general research and improve SCD prevention.
Collapse
Affiliation(s)
- Krzysztof Badura
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Dominika Buławska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Bartłomiej Dąbek
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Alicja Witkowska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Wiktoria Lisińska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Ewa Radzioch
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Sylwia Skwira
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| |
Collapse
|
11
|
Semino F, Darche FF, Bruehl C, Koenen M, Skladny H, Katus HA, Frey N, Draguhn A, Schweizer PA. GPD1L-A306del modifies sodium current in a family carrying the dysfunctional SCN5A-G1661R mutation associated with Brugada syndrome. Pflugers Arch 2024; 476:229-242. [PMID: 38036776 DOI: 10.1007/s00424-023-02882-0] [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: 04/26/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023]
Abstract
Loss-of-function variants of SCN5A, encoding the sodium channel alpha subunit Nav1.5 are associated with high phenotypic variability and multiple cardiac presentations, while underlying mechanisms are incompletely understood. Here we investigated a family with individuals affected by Brugada Syndrome (BrS) of different severity and aimed to unravel the underlying genetic and electrophysiological basis.Next-generation sequencing was used to identify the genetic variants carried by family members. The index patient, who was severely affected by arrhythmogenic BrS, carried previously uncharacterized variants of Nav1.5 (SCN5A-G1661R) and glycerol-3-phosphate dehydrogenase-1-like protein (GPD1L-A306del) in a double heterozygous conformation. Family members exclusively carrying SCN5A-G1661R showed asymptomatic Brugada ECG patterns, while another patient solely carrying GPD1L-A306del lacked any clinical phenotype.To assess functional mechanisms, Nav1.5 channels were transiently expressed in HEK-293 cells in the presence and absence of GPD1L. Whole-cell patch-clamp recordings revealed loss of sodium currents after homozygous expression of SCN5A-G1661R, and reduction of current amplitude to ~ 50% in cells transfected with equal amounts of wildtype and mutant Nav1.5. Co-expression of wildtype Nav1.5 and GPD1L showed a trend towards increased sodium current amplitudes and a hyperpolarizing shift in steady-state activation and -inactivation compared to sole SCN5A expression. Application of the GPD1L-A306del variant shifted steady-state activation to more hyperpolarized and inactivation to more depolarized potentials.In conclusion, SCN5A-G1661R produces dysfunctional channels and associates with BrS. SCN5A mediated currents are modulated by co-expression of GDP1L and this interaction is altered by mutations in both proteins. Thus, additive genetic burden may aggravate disease severity, explaining higher arrhythmogenicity in double mutation carriers.
Collapse
Affiliation(s)
- Francesca Semino
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Fabrice F Darche
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany
| | - Claus Bruehl
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Michael Koenen
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Heyko Skladny
- SYNLAB MVZ Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Andreas Draguhn
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Patrick A Schweizer
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
12
|
Qi M, Ma S, Liu J, Liu X, Wei J, Lu WJ, Zhang S, Chang Y, Zhang Y, Zhong K, Yan Y, Zhu M, Song Y, Chen Y, Hao G, Wang J, Wang L, Lee AS, Chen X, Wang Y, Lan F. In Vivo Base Editing of Scn5a Rescues Type 3 Long QT Syndrome in Mice. Circulation 2024; 149:317-329. [PMID: 37965733 DOI: 10.1161/circulationaha.123.065624] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/17/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Pathogenic variants in SCN5A can result in long QT syndrome type 3, a life-threatening genetic disease. Adenine base editors can convert targeted A T base pairs to G C base pairs, offering a promising tool to correct pathogenic variants. METHODS We generated a long QT syndrome type 3 mouse model by introducing the T1307M pathogenic variant into the Scn5a gene. The adenine base editor was split into 2 smaller parts and delivered into the heart by adeno-associated virus serotype 9 (AAV9-ABEmax) to correct the T1307M pathogenic variant. RESULTS Both homozygous and heterozygous T1307M mice showed significant QT prolongation. Carbachol administration induced Torsades de Pointes or ventricular tachycardia for homozygous T1307M mice (20%) but not for heterozygous or wild-type mice. A single intraperitoneal injection of AAV9-ABEmax at postnatal day 14 resulted in up to 99.20% Scn5a transcripts corrected in T1307M mice. Scn5a mRNA correction rate >60% eliminated QT prolongation; Scn5a mRNA correction rate <60% alleviated QT prolongation. Partial Scn5a correction resulted in cardiomyocytes heterogeneity, which did not induce severe arrhythmias. We did not detect off-target DNA or RNA editing events in ABEmax-treated mouse hearts. CONCLUSIONS These findings show that in vivo AAV9-ABEmax editing can correct the variant Scn5a allele, effectively ameliorating arrhythmia phenotypes. Our results offer a proof of concept for the treatment of hereditary arrhythmias.
Collapse
Affiliation(s)
- Man Qi
- Shenzhen Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Fuwai Hospital, Shenzhen, China (M.Q., S.M., X.L., Y. Chang, Y.Z., Y.Y., M.Z., L.W.)
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
- Chinese PLA General Hospital, College of Pulmonary & Critical Care Medicine, Beijing Key Laboratory of OTIR, Beijing, China (M.Q.)
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China (M.Q., Y. Chen)
| | - Shuhong Ma
- Shenzhen Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Fuwai Hospital, Shenzhen, China (M.Q., S.M., X.L., Y. Chang, Y.Z., Y.Y., M.Z., L.W.)
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
| | - Jingtong Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China (J.L., Y.W.)
| | - Xujie Liu
- Shenzhen Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Fuwai Hospital, Shenzhen, China (M.Q., S.M., X.L., Y. Chang, Y.Z., Y.Y., M.Z., L.W.)
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, China (X.L., F.L.)
| | - Jingjing Wei
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
| | - Wen-Jing Lu
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (W.-J.L., S.Z., F.L.)
| | - Siyao Zhang
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (W.-J.L., S.Z., F.L.)
| | - Yun Chang
- Shenzhen Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Fuwai Hospital, Shenzhen, China (M.Q., S.M., X.L., Y. Chang, Y.Z., Y.Y., M.Z., L.W.)
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
| | - Yongshuai Zhang
- Shenzhen Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Fuwai Hospital, Shenzhen, China (M.Q., S.M., X.L., Y. Chang, Y.Z., Y.Y., M.Z., L.W.)
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
| | - Kejia Zhong
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
| | - Yuting Yan
- Shenzhen Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Fuwai Hospital, Shenzhen, China (M.Q., S.M., X.L., Y. Chang, Y.Z., Y.Y., M.Z., L.W.)
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
| | - Min Zhu
- Shenzhen Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Fuwai Hospital, Shenzhen, China (M.Q., S.M., X.L., Y. Chang, Y.Z., Y.Y., M.Z., L.W.)
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
| | - Yabing Song
- School of Life Sciences, Tsinghua University, Beijing, China (Y.S., J. Wang)
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China (M.Q., Y. Chen)
| | - Guoliang Hao
- Henan Academy of Innovations in Medical Science, Zhengzhou, China (G.H.)
| | - Jianbin Wang
- School of Life Sciences, Tsinghua University, Beijing, China (Y.S., J. Wang)
| | - Li Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Fuwai Hospital, Shenzhen, China (M.Q., S.M., X.L., Y. Chang, Y.Z., Y.Y., M.Z., L.W.)
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
| | - Andrew S Lee
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China (A.S.L.)
| | - Xiangbo Chen
- Hangzhou Rongze Biotechnology Group Co, Ltd, Hangzhou, China (X.C.)
| | - Yongming Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China (J.L., Y.W.)
| | - Feng Lan
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China (M.Q., S.M., X.L., J. Wei, Y. Chang, Y.Z., K.Z., Y.Y., M.Z., L.W., F.L.)
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, China (X.L., F.L.)
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (W.-J.L., S.Z., F.L.)
| |
Collapse
|
13
|
Ma JG, Vandenberg JI, Ng CA. Development of automated patch clamp assays to overcome the burden of variants of uncertain significance in inheritable arrhythmia syndromes. Front Physiol 2023; 14:1294741. [PMID: 38089476 PMCID: PMC10712320 DOI: 10.3389/fphys.2023.1294741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 10/16/2024] Open
Abstract
Advances in next-generation sequencing have been exceptionally valuable for identifying variants in medically actionable genes. However, for most missense variants there is insufficient evidence to permit definitive classification of variants as benign or pathogenic. To overcome the deluge of Variants of Uncertain Significance, there is an urgent need for high throughput functional assays to assist with the classification of variants. Advances in parallel planar patch clamp technologies has enabled the development of automated high throughput platforms capable of increasing throughput 10- to 100-fold compared to manual patch clamp methods. Automated patch clamp electrophysiology is poised to revolutionize the field of functional genomics for inheritable cardiac ion channelopathies. In this review, we outline i) the evolution of patch clamping, ii) the development of high-throughput automated patch clamp assays to assess cardiac ion channel variants, iii) clinical application of these assays and iv) where the field is heading.
Collapse
Affiliation(s)
- Joanne G. Ma
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Jamie I. Vandenberg
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Chai-Ann Ng
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
14
|
Moore OM, Dorn LE, Wehrens XHT. Variant-specific therapy for long QT syndrome type 3. Heart Rhythm 2023; 20:718-719. [PMID: 36806575 PMCID: PMC10913134 DOI: 10.1016/j.hrthm.2023.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Affiliation(s)
- Oliver M Moore
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas; Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas; Department of Neuroscience, Baylor College of Medicine, Houston, Texas
| | - Lauren E Dorn
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas; Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas; Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas; Department of Neuroscience, Baylor College of Medicine, Houston, Texas; Department of Medicine, Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Center for Space Medicine, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
15
|
Yan Z, Zhong L, Zhu W, Chung SK, Hou P. Chinese herbal medicine for the treatment of cardiovascular diseases ─ targeting cardiac ion channels. Pharmacol Res 2023; 192:106765. [PMID: 37075871 DOI: 10.1016/j.phrs.2023.106765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of morbidity and mortality, imposing an increasing global health burden. Cardiac ion channels (voltage-gated NaV, CaV, KVs, and others) synergistically shape the cardiac action potential (AP) and control the heartbeat. Dysfunction of these channels, due to genetic mutations, transcriptional or post-translational modifications, may disturb the AP and lead to arrhythmia, a major risk for CVD patients. Although there are five classes of anti-arrhythmic drugs available, they can have varying levels of efficacies and side effects on patients, possibly due to the complex pathogenesis of arrhythmias. As an alternative treatment option, Chinese herbal remedies have shown promise in regulating cardiac ion channels and providing anti-arrhythmic effects. In this review, we first discuss the role of cardiac ion channels in maintaining normal heart function and the pathogenesis of CVD, then summarize the classification of Chinese herbal compounds, and elaborate detailed mechanisms of their efficacy in regulating cardiac ion channels and in alleviating arrhythmia and CVD. We also address current limitations and opportunities for developing new anti-CVD drugs based on Chinese herbal medicines.
Collapse
Affiliation(s)
- Zhenzhen Yan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Ling Zhong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Wandi Zhu
- Cardiovascular Medicine Division and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Sookja Kim Chung
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China; Faculty of Medicine & Faculty of Innovation Engineering at Macau University of Science and Technology, Taipa, Macao SAR, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Panpan Hou
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute. Zhuhai, Guangdong, China.
| |
Collapse
|
16
|
Long W, Li S, He Y, Lin J, Li M, Wen Z. Unraveling Structural Alerts in Marketed Drugs for Improving Adverse Outcome Pathway Framework of Drug-Induced QT Prolongation. Int J Mol Sci 2023; 24:ijms24076771. [PMID: 37047744 PMCID: PMC10095420 DOI: 10.3390/ijms24076771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
In pharmaceutical treatment, many non-cardiac drugs carry the risk of prolonging the QT interval, which can lead to fatal cardiac complications such as torsades de points (TdP). Although the unexpected blockade of ion channels has been widely considered to be one of the main reasons for affecting the repolarization phase of the cardiac action potential and leading to QT interval prolongation, the lack of knowledge regarding chemical structures in drugs that may induce the prolongation of the QT interval remains a barrier to further understanding the underlying mechanism and developing an effective prediction strategy. In this study, we thoroughly investigated the differences in chemical structures between QT-prolonging drugs and drugs with no drug-induced QT prolongation (DIQT) concerns, based on the Drug-Induced QT Prolongation Atlas (DIQTA) dataset. Three categories of structural alerts (SAs), namely amines, ethers, and aromatic compounds, appeared in large quantities in QT-prolonging drugs, but rarely in drugs with no DIQT concerns, indicating a close association between SAs and the risk of DIQT. Moreover, using the molecular descriptors associated with these three categories of SAs as features, the structure–activity relationship (SAR) model for predicting the high risk of inducing QT interval prolongation of marketed drugs achieved recall rates of 72.5% and 80.0% for the DIQTA dataset and the FDA Adverse Event Reporting System (FAERS) dataset, respectively. Our findings may promote a better understanding of the mechanism of DIQT and facilitate research on cardiac adverse drug reactions in drug development.
Collapse
Affiliation(s)
- Wulin Long
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Shihai Li
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Yujie He
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Jinzhu Lin
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Menglong Li
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Zhining Wen
- College of Chemistry, Sichuan University, Chengdu 610064, China
- Medical Big Data Center, Sichuan University, Chengdu 610064, China
| |
Collapse
|
17
|
Boßelmann CM, Hedrich UBS, Lerche H, Pfeifer N. Predicting functional effects of ion channel variants using new phenotypic machine learning methods. PLoS Comput Biol 2023; 19:e1010959. [PMID: 36877742 PMCID: PMC10019634 DOI: 10.1371/journal.pcbi.1010959] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/16/2023] [Accepted: 02/19/2023] [Indexed: 03/07/2023] Open
Abstract
Missense variants in genes encoding ion channels are associated with a spectrum of severe diseases. Variant effects on biophysical function correlate with clinical features and can be categorized as gain- or loss-of-function. This information enables a timely diagnosis, facilitates precision therapy, and guides prognosis. Functional characterization presents a bottleneck in translational medicine. Machine learning models may be able to rapidly generate supporting evidence by predicting variant functional effects. Here, we describe a multi-task multi-kernel learning framework capable of harmonizing functional results and structural information with clinical phenotypes. This novel approach extends the human phenotype ontology towards kernel-based supervised machine learning. Our gain- or loss-of-function classifier achieves high performance (mean accuracy 0.853 SD 0.016, mean AU-ROC 0.912 SD 0.025), outperforming both conventional baseline and state-of-the-art methods. Performance is robust across different phenotypic similarity measures and largely insensitive to phenotypic noise or sparsity. Localized multi-kernel learning offered biological insight and interpretability by highlighting channels with implicit genotype-phenotype correlations or latent task similarity for downstream analysis.
Collapse
Affiliation(s)
- Christian Malte Boßelmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
- Methods in Medical Informatics, Department of Computer Science, University of Tuebingen, Tuebingen, Germany
| | - Ulrike B. S. Hedrich
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
- * E-mail: (HL); (NP)
| | - Nico Pfeifer
- Methods in Medical Informatics, Department of Computer Science, University of Tuebingen, Tuebingen, Germany
- Institute for Bioinformatics and Medical Informatics, University of Tuebingen, Tuebingen, Germany
- * E-mail: (HL); (NP)
| |
Collapse
|
18
|
Specterman MJ, Behr ER. Cardiogenetics: the role of genetic testing for inherited arrhythmia syndromes and sudden death. Heart 2023; 109:434-441. [PMID: 36167638 DOI: 10.1136/heartjnl-2021-320015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/06/2022] [Indexed: 12/07/2022] Open
Abstract
There have been remarkable advances in our knowledge of the underlying heritability of cardiac arrhythmias. Long QT syndrome, Brugada syndrome, catecholaminergic polymorphic ventricular tachycardia, progressive cardiac conduction disease and the short QT syndrome comprise the inherited arrhythmia syndromes (IASs). Pathogenic variants in cardiac ion channel and calcium handling protein genes lead to these conditions, usually in the absence of overt structural cardiac disease. Diagnosis is contingent on the ECG phenotype but genetic testing may help to confirm the diagnosis and provide information on the mechanism of arrhythmogenesis that may guide treatment and provide prognostic information in relation to the risk of sudden arrhythmic death. Clinical genetic testing uses 'panels' of genes that are the likely culprits for the IASs being investigated. An International Consortium (Clinical Genome Resource) has curated gene panels based on genetic and experimental evidence of causation of inherited conditions and that have a role in clinical genetic testing. A 'single gene' or monogenic basis for IASs exists but in future, missing heritability and incomplete penetrance will be uncovered by association of common variants through genome-wide association studies. Novel rare variants will also be detected through whole-genome sequencing. The formulation of polygenic risk scores will likely help to predict phenotypic expression and response to treatments/risk stratification and move genetic testing very much to the fore of the diagnostic process.
Collapse
Affiliation(s)
- Mark J Specterman
- Cardiovascular Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Elijah R Behr
- Cardiovascular Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| |
Collapse
|
19
|
Miller JA, Moise N, Weinberg SH. Modeling incomplete penetrance in long QT syndrome type 3 through ion channel heterogeneity: an in silico population study. Am J Physiol Heart Circ Physiol 2023; 324:H179-H197. [PMID: 36487185 PMCID: PMC10072004 DOI: 10.1152/ajpheart.00430.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Many cardiac diseases are characterized by an increased late sodium current, including heart failure, hypertrophic cardiomyopathy, and inherited long QT syndrome type 3 (LQT3). The late sodium current in LQT3 is caused by a gain-of-function mutation in the voltage-gated sodium channel Nav1.5. Despite a well-defined genetic cause of LQT3, treatment remains inconsistent because of incomplete penetrance of the mutation and variability of antiarrhythmic efficacy. Here, we investigate the relationship between LQT3-associated mutation incomplete penetrance and variability in ion channel expression, simulating a population of 1,000 individuals with the O'Hara-Rudy model of the human ventricular myocyte. We first simulate healthy electrical activity (i.e., in the absence of a mutation) and then incorporate heterozygous expression for three LQT3-associated mutations (Y1795C, I1768V, and ΔKPQ), to directly compare the effects of each mutation on individuals across a diverse population. For all mutations, we find that susceptibility, defined by either the presence of an early afterdepolarization (EAD) or prolonged action potential duration (APD), primarily depends on the balance between the conductance of IKr and INa, for which individuals with a higher IKr-to-INa ratio are less susceptible. Furthermore, we find distinct differences across the population, observing individuals susceptible to zero, one, two, or all three mutations. Individuals tend to be less susceptible with an appropriate balance of repolarizing currents, typically via increased IKs or IK1. Interestingly, the more critical repolarizing current is mutation specific. We conclude that the balance between key currents plays a significant role in mutant-specific presentation of the disease phenotype in LQT3.NEW & NOTEWORTHY An in silico population approach investigates the relationship between variability in ion channel expression and gain-of-function mutations in the voltage-gated sodium channel associated with the congenital disorder long QT syndrome type 3 (LQT3). We find that ion channel variability can contribute to incomplete penetrance of the mutation, with mutant-specific differences in ion channel conductances leading to susceptibility to proarrhythmic action potential duration prolongation or early afterdepolarizations.
Collapse
Affiliation(s)
- Jacob A Miller
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Nicolae Moise
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Seth H Weinberg
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
20
|
Du P, Joshi V, Beyder A. Tracking Gut Motility in Organ and Cultures. Methods Mol Biol 2023; 2644:449-466. [PMID: 37142940 DOI: 10.1007/978-1-0716-3052-5_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Gastrointestinal (GI) motility is a key component of digestive health, and it is complex, involving a multitude of cell types and mechanisms to drive both rhythmic and arrhythmic activity. Tracking GI motility in organ and tissue cultures across multiple temporal (seconds, minutes, hours, days) scales can provide valuable information regarding dysmotility and to evaluate treatment options. Here, the chapter describes a simple method to monitor GI motility in organotypic cultures, using a single video camera is placed perpendicularly to the surface of the tissue. A cross-correlational analysis is used to track the relative movements of tissues between subsequent frames and subsequent fitting procedures to fit finite element functions to the deformed tissue to calculate the strain fields. Additional motility index measures from the displacement information are used to further quantify the behaviors of the tissues that are maintained in organotypic culture over days. The protocols presented in this chapter can be adapted to study organotypic cultures from other organs.
Collapse
Affiliation(s)
- Peng Du
- Auckland Bioengineering Institute, Department of Engineering Science and Biomedical Engineering, University of Auckland, Auckland, New Zealand.
| | - Vikram Joshi
- Department of Physiology and Biomedical Engineering, Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Arthur Beyder
- Department of Physiology and Biomedical Engineering, Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
21
|
Brunklaus A, Feng T, Brünger T, Perez-Palma E, Heyne H, Matthews E, Semsarian C, Symonds JD, Zuberi SM, Lal D, Schorge S. Gene variant effects across sodium channelopathies predict function and guide precision therapy. Brain 2022; 145:4275-4286. [PMID: 35037686 PMCID: PMC9897196 DOI: 10.1093/brain/awac006] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/27/2021] [Accepted: 12/10/2021] [Indexed: 11/14/2022] Open
Abstract
Pathogenic variants in the voltage-gated sodium channel gene family lead to early onset epilepsies, neurodevelopmental disorders, skeletal muscle channelopathies, peripheral neuropathies and cardiac arrhythmias. Disease-associated variants have diverse functional effects ranging from complete loss-of-function to marked gain-of-function. Therapeutic strategy is likely to depend on functional effect. Experimental studies offer important insights into channel function but are resource intensive and only performed in a minority of cases. Given the evolutionarily conserved nature of the sodium channel genes, we investigated whether similarities in biophysical properties between different voltage-gated sodium channels can predict function and inform precision treatment across sodium channelopathies. We performed a systematic literature search identifying functionally assessed variants in any of the nine voltage-gated sodium channel genes until 28 April 2021. We included missense variants that had been electrophysiologically characterized in mammalian cells in whole-cell patch-clamp recordings. We performed an alignment of linear protein sequences of all sodium channel genes and correlated variants by their overall functional effect on biophysical properties. Of 951 identified records, 437 sodium channel-variants met our inclusion criteria and were reviewed for functional properties. Of these, 141 variants were epilepsy-associated (SCN1/2/3/8A), 79 had a neuromuscular phenotype (SCN4/9/10/11A), 149 were associated with a cardiac phenotype (SCN5/10A) and 68 (16%) were considered benign. We detected 38 missense variant pairs with an identical disease-associated variant in a different sodium channel gene. Thirty-five out of 38 of those pairs resulted in similar functional consequences, indicating up to 92% biophysical agreement between corresponding sodium channel variants (odds ratio = 11.3; 95% confidence interval = 2.8 to 66.9; P < 0.001). Pathogenic missense variants were clustered in specific functional domains, whereas population variants were significantly more frequent across non-conserved domains (odds ratio = 18.6; 95% confidence interval = 10.9-34.4; P < 0.001). Pore-loop regions were frequently associated with loss-of-function variants, whereas inactivation sites were associated with gain-of-function (odds ratio = 42.1, 95% confidence interval = 14.5-122.4; P < 0.001), whilst variants occurring in voltage-sensing regions comprised a range of gain- and loss-of-function effects. Our findings suggest that biophysical characterisation of variants in one SCN-gene can predict channel function across different SCN-genes where experimental data are not available. The collected data represent the first gain- versus loss-of-function topological map of SCN proteins indicating shared patterns of biophysical effects aiding variant analysis and guiding precision therapy. We integrated our findings into a free online webtool to facilitate functional sodium channel gene variant interpretation (http://SCN-viewer.broadinstitute.org).
Collapse
Affiliation(s)
- Andreas Brunklaus
- Correspondence to: Dr Andreas Brunklaus, MD Fraser of Allander Neurosciences Unit Office Block, Ground Floor, Zone 2 Royal Hospital for Children 1345 Govan Road Glasgow G51 4TF, UK E-mail:
| | | | | | - Eduardo Perez-Palma
- Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Henrike Heyne
- Genomic and Personalized Medicine, Digital Health Center, Hasso Plattner Institute, Potsdam, Germany
- Hasso Plattner Institute, Mount Sinai School of Medicine, New York, NY, USA
- Institute for Molecular Medicine Finland: FIMM, Helsinki, Finland
| | - Emma Matthews
- Atkinson Morley Neuromuscular Centre, St George’s University Hospitals NHS Foundation Trust, London, UK
- Molecular and Clinical Sciences Research Institute, St George’s University of London, London, UK
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, Australia
- Sydney Medical School Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Joseph D Symonds
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Glasgow, UK
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Sameer M Zuberi
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Glasgow, UK
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Dennis Lal
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, USA
- Stanley Center for Psychiatric Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stephanie Schorge
- Correspondence may also be addressed to: Professor Stephanie Schorge, PhD Department of Neuroscience Physiology and Pharmacology UCL, London WC1E 6BT, UK E-mail:
| |
Collapse
|
22
|
Cardiotoxicity of pyrethroids: molecular mechanisms and therapeutic options for acute and long-term toxicity. Biochem Soc Trans 2022; 50:1737-1751. [PMID: 36383062 DOI: 10.1042/bst20220593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022]
Abstract
Pyrethroids (PY) are synthetic pesticides used in many applications ranging from large-scale agriculture to household maintenance. Their classical mechanisms of action are associated with binding to the sodium channel of insect neurons, disrupting its inactivation, ensuring their use as insecticides. However, PY can also lead to toxicity in vertebrates, including humans. In most toxicological studies, the impact of PY on heart function is neglected. Acute exposure to a high dose of PY causes enhancement of the late sodium current (INaL), which impairs the action potential waveform and can cause severe cardiac arrhythmias. Moreover, long-term, low-dose exposure to PY displays oxidative stress in the heart, which could induce tissue remodeling and impairment. Isolated and preliminary evidence supports that, for acute exposure to PY, an antiarrhythmic therapy with ranolazine (an INaL blocker), can be a promising therapeutic approach. Besides, heart tissue remodeling associated with low doses and long-term exposure to PY seems to benefit from antioxidant therapy. Despite significant leaps in understanding the mechanical details of PY intoxication, currently, few studies are focusing on the heart. In this review, we present what is known and what are the gaps in the field of cardiotoxicity induced by PY.
Collapse
|
23
|
Comollo TW, Zou X, Zhang C, Kesters D, Hof T, Sampson KJ, Kass RS. Exploring mutation specific beta blocker pharmacology of the pathogenic late sodium channel current from patient-specific pluripotent stem cell myocytes derived from long QT syndrome mutation carriers. Channels (Austin) 2022; 16:173-184. [PMID: 35949058 PMCID: PMC9373745 DOI: 10.1080/19336950.2022.2106025] [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] [Indexed: 01/31/2023] Open
Abstract
The congenital long QT syndrome (LQTS), one of the most common cardiac channelopathies, is characterized by delayed ventricular repolarization underlying prolongation of the QT interval of the surface electrocardiogram. LQTS is caused by mutations in genes coding for cardiac ion channels or ion channel-associated proteins. The major therapeutic approach to LQTS management is beta blocker therapy which has been shown to be effective in treatment of LQTS variants caused by mutations in K+ channels. However, this approach has been questioned in the treatment of patients identified as LQTS variant 3(LQT3) patients who carry mutations in SCN5A, the gene coding for the principal cardiac Na+ channel. LQT3 mutations are gain of function mutations that disrupt spontaneous Na+ channel inactivation and promote persistent or late Na+ channel current (INaL) that delays repolarization and underlies QT prolongation. Clinical investigation of patients with the two most common LQT3 mutations, the ΔKPQ and the E1784K mutations, found beta blocker treatment a useful therapeutic approach for managing arrhythmias in this patient population. However, there is little experimental data that reveals the mechanisms underlying these antiarrhythmic actions. Here, we have investigated the effects of the beta blocker propranolol on INaL expressed by ΔKPQ and E1784K channels in induced pluripotent stem cells derived from patients carrying these mutations. Our results indicate that propranolol preferentially inhibits INaL expressed by these channels suggesting that the protective effects of propranolol in treating LQT3 patients is due in part to modulation of INaL.
Collapse
Affiliation(s)
- Thomas W. Comollo
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Xinle Zou
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Chuangeng Zhang
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Divya Kesters
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Thomas Hof
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Kevin J. Sampson
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA
| | - Robert S. Kass
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia, NY, USA,CONTACT Robert S. Kass
| |
Collapse
|
24
|
Gamal El-Din TM. When the Gates Swing Open Only: Arrhythmia Mutations That Target the Fast Inactivation Gate of Na v1.5. Cells 2022; 11:cells11233714. [PMID: 36496974 PMCID: PMC9735811 DOI: 10.3390/cells11233714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Nav1.5 is the main voltage-gated sodium channel found in cardiac muscle, where it facilitates the fast influx of Na+ ions across the cell membrane, resulting in the fast depolarization phase-phase 0 of the cardiac action potential. As a result, it plays a major role in determining the amplitude and the upstroke velocity of the cardiac impulse. Quantitively, cardiac sodium channel activates in less than a millisecond to trigger the cardiac action potential and inactivates within 2-3 ms to facilitate repolarization and return to the resting state in preparation for firing the next action potential. Missense mutations in the gene that encodes Nav1.5 (SCN5A), change these time constants which leads to a wide spectrum of cardiac diseases ranging from long QT syndrome type 3 (LQT3) to sudden cardiac death. In this mini-review I will focus on the missense mutations in the inactivation gate of Nav1.5 that results in arrhythmia, attempting to correlate the location of the missense mutation to their specific phenotype.
Collapse
|
25
|
Qauli AI, Yoo Y, Marcellinus A, Lim KM. Verification of the Efficacy of Mexiletine Treatment for the A1656D Mutation on Downgrading Reentrant Tachycardia Using a 3D Cardiac Electrophysiological Model. Bioengineering (Basel) 2022; 9:531. [PMID: 36290499 PMCID: PMC9598628 DOI: 10.3390/bioengineering9100531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 10/21/2023] Open
Abstract
The SCN5A mutations have been long associated with long QT variant 3 (LQT3). Recent experimental and computation studies have reported that mexiletine effectively treats LQT3 patients associated with the A1656D mutation. However, they have primarily focused on cellular level evaluations and have only looked at the effects of mexiletine on action potential duration (APD) or QT interval reduction. We further investigated mexiletine's effects on cardiac cells through simulations of single-cell (behavior of alternant occurrence) and 3D (with and without mexiletine). We discovered that mexiletine could shorten the cell's APD and change the alternant's occurrence to a shorter basic cycle length (BCL) between 350 and 420 ms. The alternant also appeared at a normal heart rate under the A1656D mutation. Furthermore, the 3D ventricle simulations revealed that mexiletine could reduce the likelihood of a greater spiral wave breakup in the A1656D mutant condition by minimizing the appearance of rotors. In conclusion, we found that mexiletine could provide extra safety features during therapy for LQT3 patients because it can change the alternant occurrence from a normal to a faster heart rate, and it reduces the chance of a spiral wave breakup. Therefore, these findings emphasize the promising efficacy of mexiletine in treating LQT3 patients under the A1656D mutation.
Collapse
Affiliation(s)
- Ali Ikhsanul Qauli
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
- Robotics and Artificial Intelligence Engineering, Faculty of Advanced Technology and Multidiscipline, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia
| | - Yedam Yoo
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
| | - Aroli Marcellinus
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
| | - Ki Moo Lim
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
| |
Collapse
|
26
|
Postrigan AE, Babushkina NP, Svintsova LI, Plotnikova IV, Skryabin NA. Clinical and Genetic Characteristics of Congenital Long QT Syndrome. RUSS J GENET+ 2022. [DOI: 10.1134/s1022795422100064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
27
|
vom Dahl C, Müller CE, Berisha X, Nagel G, Zimmer T. Coupling the Cardiac Voltage-Gated Sodium Channel to Channelrhodopsin-2 Generates Novel Optical Switches for Action Potential Studies. MEMBRANES 2022; 12:907. [PMID: 36295666 PMCID: PMC9607247 DOI: 10.3390/membranes12100907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 06/16/2023]
Abstract
Voltage-gated sodium (Na+) channels respond to short membrane depolarization with conformational changes leading to pore opening, Na+ influx, and action potential (AP) upstroke. In the present study, we coupled channelrhodopsin-2 (ChR2), the key ion channel in optogenetics, directly to the cardiac voltage-gated Na+ channel (Nav1.5). Fusion constructs were expressed in Xenopus laevis oocytes, and electrophysiological recordings were performed by the two-microelectrode technique. Heteromeric channels retained both typical Nav1.5 kinetics and light-sensitive ChR2 properties. Switching to the current-clamp mode and applying short blue-light pulses resulted either in subthreshold depolarization or in a rapid change of membrane polarity typically seen in APs of excitable cells. To study the effect of individual K+ channels on the AP shape, we co-expressed either Kv1.2 or hERG with one of the Nav1.5-ChR2 fusions. As expected, both delayed rectifier K+ channels shortened AP duration significantly. Kv1.2 currents remarkably accelerated initial repolarization, whereas hERG channel activity efficiently restored the resting membrane potential. Finally, we investigated the effect of the LQT3 deletion mutant ΔKPQ on the AP shape and noticed an extremely prolonged AP duration that was directly correlated to the size of the non-inactivating Na+ current fraction. In conclusion, coupling of ChR2 to a voltage-gated Na+ channel generates optical switches that are useful for studying the effect of individual ion channels on the AP shape. Moreover, our novel optogenetic approach provides the potential for an application in pharmacology and optogenetic tissue-engineering.
Collapse
Affiliation(s)
- Christian vom Dahl
- Institute of Physiology II, University Hospital Jena, Friedrich Schiller University, 07740 Jena, Germany
| | - Christoph Emanuel Müller
- Institute of Physiology II, University Hospital Jena, Friedrich Schiller University, 07740 Jena, Germany
| | - Xhevat Berisha
- Institute of Physiology II, University Hospital Jena, Friedrich Schiller University, 07740 Jena, Germany
| | - Georg Nagel
- Institute of Physiology—Neurophysiology, Julius Maximilians University, 97070 Wuerzburg, Germany
| | - Thomas Zimmer
- Institute of Physiology II, University Hospital Jena, Friedrich Schiller University, 07740 Jena, Germany
| |
Collapse
|
28
|
Katsuta E, Takabe K, Vujcic M, Gottlieb PA, Dai T, Mercado-Perez A, Beyder A, Wang Q, Opyrchal M. Mechano-Sensing Channel PIEZO2 Enhances Invasive Phenotype in Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:9909. [PMID: 36077309 PMCID: PMC9455988 DOI: 10.3390/ijms23179909] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Mechanically gated PIEZO channels lead to an influx of cations, activation of additional Ca2+ channels, and cell depolarization. This study aimed to investigate PIEZO2's role in breast cancer. METHODS The clinical relevance of PIEZO2 expression in breast cancer patient was analyzed in a publicly available dataset. Utilizing PIEZO2 overexpressed breast cancer cells, and in vitro and in vivo experiments were conducted. RESULTS High expression of PIEZO2 was correlated with a worse survival in triple-negative breast cancer (TNBC) but not in other subtypes. Increased PEIZO2 channel function was confirmed in PIEZO2 overexpressed cells after mechanical stimulation. PIEZO2 overexpressed cells showed increased motility and invasive phenotypes as well as higher expression of SNAIL and Vimentin and lower expression of E-cadherin in TNBC cells. Correspondingly, high expression of PIEZO2 was correlated with the increased expression of epithelial-mesenchymal transition (EMT)-related genes in a TNBC patient. Activated Akt signaling was observed in PIEZO2 overexpressed TNBC cells. PIEZO2 overexpressed MDA-MB-231 cells formed a significantly higher number of lung metastases after orthotopic implantation. CONCLUSION PIEZO2 activation led to enhanced SNAIL stabilization through Akt activation. It enhanced Vimentin and repressed E-cadherin transcription, resulting in increased metastatic potential and poor clinical outcomes in TNBC patients.
Collapse
Affiliation(s)
- Eriko Katsuta
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY 14203, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
- Department of Surgery, Yokohama City University, Yokohama 236-0004, Japan
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
- Department of Breast Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Marija Vujcic
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Philip A. Gottlieb
- Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Tao Dai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Arnaldo Mercado-Perez
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Qingfei Wang
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mateusz Opyrchal
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
29
|
Rieder M, Kreifels P, Stuplich J, Ziupa D, Servatius H, Nicolai L, Castiglione A, Zweier C, Asatryan B, Odening KE. Genotype-Specific ECG-Based Risk Stratification Approaches in Patients With Long-QT Syndrome. Front Cardiovasc Med 2022; 9:916036. [PMID: 35911527 PMCID: PMC9329832 DOI: 10.3389/fcvm.2022.916036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background Congenital long-QT syndrome (LQTS) is a major cause of sudden cardiac death (SCD) in young individuals, calling for sophisticated risk assessment. Risk stratification, however, is challenging as the individual arrhythmic risk varies pronouncedly, even in individuals carrying the same variant. Materials and Methods In this study, we aimed to assess the association of different electrical parameters with the genotype and the symptoms in patients with LQTS. In addition to the heart-rate corrected QT interval (QTc), markers for regional electrical heterogeneity, such as QT dispersion (QTmax-QTmin in all ECG leads) and delta Tpeak/end (Tpeak/end V5 – Tpeak/end V2), were assessed in the 12-lead ECG at rest and during exercise testing. Results QTc at rest was significantly longer in symptomatic than asymptomatic patients with LQT2 (493.4 ms ± 46.5 ms vs. 419.5 ms ± 28.6 ms, p = 0.004), but surprisingly not associated with symptoms in LQT1. In contrast, post-exercise QTc (minute 4 of recovery) was significantly longer in symptomatic than asymptomatic patients with LQT1 (486.5 ms ± 7.0 ms vs. 463.3 ms ± 16.3 ms, p = 0.04), while no such difference was observed in patients with LQT2. Enhanced delta Tpeak/end and QT dispersion were only associated with symptoms in LQT1 (delta Tpeak/end 19.0 ms ± 18.1 ms vs. −4.0 ms ± 4.4 ms, p = 0.02; QT-dispersion: 54.3 ms ± 10.2 ms vs. 31.4 ms ± 10.4 ms, p = 0.01), but not in LQT2. Delta Tpeak/end was particularly discriminative after exercise, where all symptomatic patients with LQT1 had positive and all asymptomatic LQT1 patients had negative values (11.8 ± 7.9 ms vs. −7.5 ± 1.7 ms, p = 0.003). Conclusion Different electrical parameters can distinguish between symptomatic and asymptomatic patients in different genetic forms of LQTS. While the classical “QTc at rest” was only associated with symptoms in LQT2, post-exercise QTc helped distinguish between symptomatic and asymptomatic patients with LQT1. Enhanced regional electrical heterogeneity was only associated with symptoms in LQT1, but not in LQT2. Our findings indicate that genotype-specific risk stratification approaches based on electrical parameters could help to optimize risk assessment in LQTS.
Collapse
Affiliation(s)
- Marina Rieder
- Translational Cardiology, Department of Cardiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Paul Kreifels
- Department of Cardiology and Angiology I, Faculty of Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Judith Stuplich
- Department of Cardiology and Angiology I, Faculty of Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - David Ziupa
- Department of Cardiology and Angiology I, Faculty of Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Helge Servatius
- Translational Cardiology, Department of Cardiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Luisa Nicolai
- Translational Cardiology, Department of Cardiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Alessandro Castiglione
- Translational Cardiology, Department of Cardiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Christiane Zweier
- Department of Human Genetics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Babken Asatryan
- Translational Cardiology, Department of Cardiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Katja E Odening
- Translational Cardiology, Department of Cardiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
30
|
Ross RL, Mavria G, Del Galdo F, Elies J. Downregulation of Vascular Hemeoxygenase-1 Leads to Vasculopathy in Systemic Sclerosis. Front Physiol 2022; 13:900631. [PMID: 35600300 PMCID: PMC9117635 DOI: 10.3389/fphys.2022.900631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a terminal disease characterized by vasculopathy, tissue fibrosis, and autoimmunity. Although the exact etiology of SSc remains unknown, endothelial dysfunction, oxidative stress, and calcium handling dysregulation have been associated with a large number of SSc-related complications such as neointima formation, vasculogenesis, pulmonary arterial hypertension, impaired angiogenesis, and cardiac arrhythmias. Hemeoxygenase-1 (HO-1) is an antioxidant enzyme involved in multiple biological actions in the cardiovascular system including vascular tone, angiogenesis, cellular proliferation, apoptosis, and oxidative stress. The aim of this work was to investigate the physiological role of HO-1 and its relevance in the cardiovascular complications occurring in SSc. We found that, in early phases of SSc, the expression of HO-1 in dermal fibroblast is lower compared to those isolated from healthy control individuals. This is particularly relevant as reduction of the HO-1/CO signaling pathway is associated with endothelial dysfunction and vasculopathy. We show evidence of the role of HO-1/carbon monoxide (CO) signaling pathway in calcium handling. Using an in vitro model of pulmonary arterial hypertension (PAH) we investigated the role of HO-1 in Ca2+ mobilization from intracellular stores. Our results indicate that HO-1 regulates calcium release from intracellular stores of human pulmonary arterial endothelial cells. We interrogated the activity of HO-1 in angiogenesis using an organotypic co-culture of fibroblast-endothelial cell. Inhibition of HO-1 significantly reduced the ability of endothelial cells to form tubules. We further investigated if this could be associated with cell motility or migration of endothelial cells into the extracellular matrix synthesized by fibroblasts. By mean of holographic imaging, we studied the morphological and functional features of endothelial cells in the presence of an HO-1 activator and selective inhibitors. Our results demonstrate that inhibition of HO-1 significantly reduces cell proliferation and cell motility (migration) of cultured endothelial cells, whilst activation of HO-1 does not modify either morphology, proliferation or motility. In addition, we investigated the actions of CO on the Kv7.1 (KCQN1) channel current, an important component of the cardiac action potential repolarization. Using electrophysiology (whole-cell patch-clamp in a recombinant system overexpressing the KCQN1 channel), we assessed the regulation of KCQN1 by CO. CORM-2, a CO donor, significantly reduced the Kv7.1 current, suggesting that HO-1/CO signaling may play a role in the modulation of the cardiac action potential via regulation of this ion channel. In summary, our results indicate a clear link between: 1) downregulation of HO-1/CO signaling; and 2) pathophysiological processes occurring in early phases of SSc, such as calcium homeostasis dysregulation, impaired angiogenesis and cardiac arrhythmias. A better understanding of the canonical actions (mainly due to the biological actions of CO), and non-canonical actions of HO-1, as well as the interaction of HO-1/CO signaling with other gasotransmitters in SSc will contribute to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Rebecca L Ross
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
- Scleroderma Programme, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, United Kingdom
| | - Georgia Mavria
- Signal Transduction and Tumour Microenvironment Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
- Scleroderma Programme, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, United Kingdom
| | - Jacobo Elies
- Cardiovascular Research Group, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
- *Correspondence: Jacobo Elies,
| |
Collapse
|
31
|
Krahn AD, Laksman Z, Sy RW, Postema PG, Ackerman MJ, Wilde AAM, Han HC. Congenital Long QT Syndrome. JACC Clin Electrophysiol 2022; 8:687-706. [PMID: 35589186 DOI: 10.1016/j.jacep.2022.02.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 12/14/2022]
Abstract
Congenital long QT syndrome (LQTS) encompasses a group of heritable conditions that are associated with cardiac repolarization dysfunction. Since its initial description in 1957, our understanding of LQTS has increased dramatically. The prevalence of LQTS is estimated to be ∼1:2,000, with a slight female predominance. The diagnosis of LQTS is based on clinical, electrocardiogram, and genetic factors. Risk stratification of patients with LQTS aims to identify those who are at increased risk of cardiac arrest or sudden cardiac death. Factors including age, sex, QTc interval, and genetic background all contribute to current risk stratification paradigms. The management of LQTS involves conservative measures such as the avoidance of QT-prolonging drugs, pharmacologic measures with nonselective β-blockers, and interventional approaches such as device therapy or left cardiac sympathetic denervation. In general, most forms of exercise are considered safe in adequately treated patients, and implantable cardioverter-defibrillator therapy is reserved for those at the highest risk. This review summarizes our current understanding of LQTS and provides clinicians with a practical approach to diagnosis and management.
Collapse
Affiliation(s)
- Andrew D Krahn
- Center for Cardiovascular Innovation, Heart Rhythm Services, Division of Cardiology, University of British Columbia, Vancouver, BC, Canada.
| | - Zachary Laksman
- Center for Cardiovascular Innovation, Heart Rhythm Services, Division of Cardiology, University of British Columbia, Vancouver, BC, Canada
| | - Raymond W Sy
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Pieter G Postema
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Michael J Ackerman
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, Minnesota, USA; Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, Minnesota, USA; Departments of Molecular Pharmacology and Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota, USA
| | - Arthur A M Wilde
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands; European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart), Academic University Medical Center, Amsterdam, the Netherlands
| | - Hui-Chen Han
- Center for Cardiovascular Innovation, Heart Rhythm Services, Division of Cardiology, University of British Columbia, Vancouver, BC, Canada; Victorian Heart Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
32
|
Chakouri N, Rivas S, Roybal D, Yang L, Diaz J, Hsu A, Mahling R, Chen BX, Owoyemi JO, DiSilvestre D, Sirabella D, Corneo B, Tomaselli GF, Dick IE, Marx SO, Ben-Johny M. Fibroblast growth factor homologous factors serve as a molecular rheostat in tuning arrhythmogenic cardiac late sodium current. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1-13. [PMID: 35662881 PMCID: PMC9161660 DOI: 10.1038/s44161-022-00060-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/04/2022] [Indexed: 05/20/2023]
Abstract
Voltage-gated sodium (Nav1.5) channels support the genesis and brisk spatial propagation of action potentials in the heart. Disruption of NaV1.5 inactivation results in a small persistent Na influx known as late Na current (I Na,L), which has emerged as a common pathogenic mechanism in both congenital and acquired cardiac arrhythmogenic syndromes. Here, using low-noise multi-channel recordings in heterologous systems, LQTS3 patient-derived iPSCs cardiomyocytes, and mouse ventricular myocytes, we demonstrate that the intracellular fibroblast growth factor homologous factors (FHF1-4) tune pathogenic I Na,L in an isoform-specific manner. This scheme suggests a complex orchestration of I Na,L in cardiomyocytes that may contribute to variable disease expressivity of NaV1.5 channelopathies. We further leverage these observations to engineer a peptide-inhibitor of I Na,L with a higher efficacy as compared to a well-established small-molecule inhibitor. Overall, these findings lend insights into molecular mechanisms underlying FHF regulation of I Na,L in pathophysiology and outline potential therapeutic avenues.
Collapse
Affiliation(s)
- Nourdine Chakouri
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Sharen Rivas
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Daniel Roybal
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Lin Yang
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
| | - Johanna Diaz
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Allen Hsu
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Ryan Mahling
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Bi-Xing Chen
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
| | | | - Deborah DiSilvestre
- Department Physiology, University of Maryland, Baltimore, MD, USA
- Division of Cardiology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Dario Sirabella
- Columbia Stem Cell Initiative, Stem Cell Core, Columbia University Irving Medical Center, NY, USA
| | - Barbara Corneo
- Columbia Stem Cell Initiative, Stem Cell Core, Columbia University Irving Medical Center, NY, USA
| | - Gordon F. Tomaselli
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
- Division of Cardiology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Ivy E. Dick
- Department Physiology, University of Maryland, Baltimore, MD, USA
| | - Steven O. Marx
- Department of Pharmacology, Columbia University, New York, NY, USA
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| |
Collapse
|
33
|
The insecticide β-Cyfluthrin induces acute arrhythmic cardiotoxicity through interaction with NaV1.5 and ranolazine reverses the phenotype. Clin Sci (Lond) 2022; 136:329-343. [PMID: 35190819 DOI: 10.1042/cs20211151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/10/2022] [Accepted: 02/22/2022] [Indexed: 11/17/2022]
Abstract
β-Cyfluthrin, a class II Pyrethroid, is an insecticide used worldwide in agriculture, horticulture (field and protected crops), viticulture, and domestic applications. β-Cyfluthrin may impair the function of biological systems; however, little information is available about its potential cardiotoxic effect. Here, we explored the acute toxicity of β-Cyfluthrin in isolated heart preparations and its cellular basis, using isolated cardiomyocytes. Moreover, β-Cyfluthrin effects on the sodium current, especially late sodium current (INa-L), were investigated using HEK-293 cells transiently expressing human NaV1.5 channels. We report that β-Cyfluthrin raised INa-L in a dose-dependent manner. β-Cyfluthrin prolonged the repolarization of the action potential and triggered oscillations on its duration. Cardiomyocytes contraction and calcium dynamics were disrupted by the pesticide with a marked incidence of non-electronic stimulated contractions. The antiarrhythmic drug Ranolazine was able to reverse most of the phenotypes observed in isolated cells. Lastly, ventricular premature beats and long QT intervals were found during β-Cyfluthrin exposure, and Ranolazine was able to attenuate them. Overall, we demonstrated that β-Cyfluthrin can cause significant cardiac alterations and Ranolazine ameliorated the phenotype. Understanding the insecticides' impacts upon electromechanical properties of the heart is important for the development of therapeutic approaches to treat cases of pesticides intoxication.
Collapse
|
34
|
Hu RM, Song EJ, Tester DJ, Deschenes I, Ackerman MJ, Makielski JC, Tan BH. Expression defect of the rare variant/Brugada mutation R1512W depends upon the SCN5A splice variant background and can be rescued by mexiletine and the common polymorphism H558R. Channels (Austin) 2021; 15:253-261. [PMID: 33535892 PMCID: PMC7872018 DOI: 10.1080/19336950.2021.1875645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 12/01/2022] Open
Abstract
Background : Mutations in SCN5A that decrease Na current underlie arrhythmia syndromes such as the Brugada syndrome (BrS). SCN5A in humans has two splice variants, one lacking a glutamine at position 1077 (Q1077del) and one containing Q1077. We investigated the effect of splice variant background on loss-of-function and rescue for R1512W, a mutation reported to cause BrS. Methods and results : We made the mutation in both variants and expressed them in HEK-293 cells for voltage-clamp study. After 24 hours of transfection, the current expression level of R1512W was reduced by ~50% in both Q1077del and Q1077 compared to the wild-type (WT) channel, respectively. The activation and inactivation midpoint were not different between WT and mutant channels in both splice variant backgrounds. However, slower time constants of recovery and enhanced intermediate inactivation were observed for R1512W/Q1077 compared with WT-Q1077, while the recovery and intermediate inactivation parameters of R1512W/Q1077del were similar to WT-Q1077del. Furthermore, both mexiletine and the common polymorphism H558R restored peak sodium current (INa) amplitude of the mutant channel by increasing the cell surface expression of SCN5A. Conclusion : These findings provide further evidence that the splice variant affects the molecular phenotype with implications for the clinical phenotype, and they provide insight into the expression defect mechanisms and potential treatment in BrS.
Collapse
Affiliation(s)
- Rou-Mu Hu
- Department of Cardiology, Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Evelyn J. Song
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David J. Tester
- Departments of Medicine, Pediatrics, and Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Isabelle Deschenes
- Department of Physiology and Cell Biology, The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, the Ohio State University, Columbus, OH, USA
| | - Michael J. Ackerman
- Departments of Medicine, Pediatrics, and Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Jonathan C. Makielski
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Bi-Hua Tan
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison, WI, USA
- Department of Physiology and Cell Biology, The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, the Ohio State University, Columbus, OH, USA
| |
Collapse
|
35
|
Rivaud MR, Delmar M, Remme CA. Heritable arrhythmia syndromes associated with abnormal cardiac sodium channel function: ionic and non-ionic mechanisms. Cardiovasc Res 2021; 116:1557-1570. [PMID: 32251506 PMCID: PMC7341171 DOI: 10.1093/cvr/cvaa082] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/01/2020] [Accepted: 04/01/2020] [Indexed: 12/19/2022] Open
Abstract
The cardiac sodium channel NaV1.5, encoded by the SCN5A gene, is responsible for the fast upstroke of the action potential. Mutations in SCN5A may cause sodium channel dysfunction by decreasing peak sodium current, which slows conduction and facilitates reentry-based arrhythmias, and by enhancing late sodium current, which prolongs the action potential and sets the stage for early afterdepolarization and arrhythmias. Yet, some NaV1.5-related disorders, in particular structural abnormalities, cannot be directly or solely explained on the basis of defective NaV1.5 expression or biophysics. An emerging concept that may explain the large disease spectrum associated with SCN5A mutations centres around the multifunctionality of the NaV1.5 complex. In this alternative view, alterations in NaV1.5 affect processes that are independent of its canonical ion-conducting role. We here propose a novel classification of NaV1.5 (dys)function, categorized into (i) direct ionic effects of sodium influx through NaV1.5 on membrane potential and consequent action potential generation, (ii) indirect ionic effects of sodium influx on intracellular homeostasis and signalling, and (iii) non-ionic effects of NaV1.5, independent of sodium influx, through interactions with macromolecular complexes within the different microdomains of the cardiomyocyte. These indirect ionic and non-ionic processes may, acting alone or in concert, contribute significantly to arrhythmogenesis. Hence, further exploration of these multifunctional effects of NaV1.5 is essential for the development of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Mathilde R Rivaud
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC (location AMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Meigberdreef 15, 1105AZ Amsterdam, The Netherlands
| | - Mario Delmar
- The Leon H. Charney Division of Cardiology, New York University School of Medicine, 435 E 30th St, NSB 707, New York, NY 10016, USA
| | - Carol Ann Remme
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC (location AMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Meigberdreef 15, 1105AZ Amsterdam, The Netherlands
| |
Collapse
|
36
|
Functional cross-talk between phosphorylation and disease-causing mutations in the cardiac sodium channel Na v1.5. Proc Natl Acad Sci U S A 2021; 118:2025320118. [PMID: 34373326 PMCID: PMC8379932 DOI: 10.1073/pnas.2025320118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The cardiac sodium channel (Nav1.5) is crucial for generating a regular heartbeat. It is thus not surprising that Nav1.5 mutations have been linked to life-threatening arrhythmias. Interestingly, Nav1.5 activity can also be altered by posttranslational modifications, such as tyrosine phosphorylation. Our combination of protein engineering and molecular modeling has revealed that the detrimental effect of a long QT3 patient mutation is only exposed when a proximal tyrosine is phosphorylated. This suggests a dynamic cross-talk between the genetic mutation and a neighboring phosphorylation, a phenomenon that could be important in other classes of proteins. Additionally, we show that phosphorylation can affect the channel’s sensitivity toward clinically relevant drugs, a finding that may prove important when devising patient-specific treatment plans. The voltage-gated sodium channel Nav1.5 initiates the cardiac action potential. Alterations of its activation and inactivation properties due to mutations can cause severe, life-threatening arrhythmias. Yet despite intensive research efforts, many functional aspects of this cardiac channel remain poorly understood. For instance, Nav1.5 undergoes extensive posttranslational modification in vivo, but the functional significance of these modifications is largely unexplored, especially under pathological conditions. This is because most conventional approaches are unable to insert metabolically stable posttranslational modification mimics, thus preventing a precise elucidation of the contribution by these modifications to channel function. Here, we overcome this limitation by using protein semisynthesis of Nav1.5 in live cells and carry out complementary molecular dynamics simulations. We introduce metabolically stable phosphorylation mimics on both wild-type (WT) and two pathogenic long-QT mutant channel backgrounds and decipher functional and pharmacological effects with unique precision. We elucidate the mechanism by which phosphorylation of Y1495 impairs steady-state inactivation in WT Nav1.5. Surprisingly, we find that while the Q1476R patient mutation does not affect inactivation on its own, it enhances the impairment of steady-state inactivation caused by phosphorylation of Y1495 through enhanced unbinding of the inactivation particle. We also show that both phosphorylation and patient mutations can impact Nav1.5 sensitivity toward the clinically used antiarrhythmic drugs quinidine and ranolazine, but not flecainide. The data highlight that functional effects of Nav1.5 phosphorylation can be dramatically amplified by patient mutations. Our work is thus likely to have implications for the interpretation of mutational phenotypes and the design of future drug regimens.
Collapse
|
37
|
Gomez‐Galeno J, Okolotowicz K, Johnson M, McKeithan WL, Mercola M, Cashman JR. Human-induced pluripotent stem cell-derived cardiomyocytes: Cardiovascular properties and metabolism and pharmacokinetics of deuterated mexiletine analogs. Pharmacol Res Perspect 2021; 9:e00828. [PMID: 34327875 PMCID: PMC8322572 DOI: 10.1002/prp2.828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 01/08/2023] Open
Abstract
Prolongation of the cardiac action potential (AP) and early after depolarizations (EADs) are electrical anomalies of cardiomyocytes that can lead to lethal arrhythmias and are potential liabilities for existing drugs and drug candidates in development. For example, long QT syndrome-3 (LQTS3) is caused by mutations in the Nav 1.5 sodium channel that debilitate channel inactivation and cause arrhythmias. We tested the hypothesis that a useful drug (i.e., mexiletine) with potential liabilities (i.e., potassium channel inhibition and adverse reactions) could be re-engineered by dynamic medicinal chemistry to afford a new drug candidate with greater efficacy and less toxicity. Human cardiomyocytes were generated from LQTS3 patient-derived induced pluripotent stem cells (hIPSCs) and normal hIPSCs to determine beneficial (on-target) and detrimental effects (off-target) of mexiletine and synthetic analogs, respectively. The approach combined "drug discovery" and "hit to lead" refinement and showed that iterations of medicinal chemistry and physiological testing afforded optimized compound 22. Compared to mexiletine, compound 22 showed a 1.85-fold greater AUC and no detectable CNS toxicity at 100 mg/kg. In vitro hepatic metabolism studies showed that 22 was metabolized via cytochrome P-450, as previously shown, and by the flavin-containing monooxygenase (FMO). Deuterated-22 showed decreased metabolism and showed acceptable cardiovascular and physicochemical properties.
Collapse
Affiliation(s)
| | - Karl Okolotowicz
- Department of MedicineCardiovascular InstituteStanford UniversityStanfordCAUSA
| | - Mark Johnson
- Human BioMolecular Research InstituteSan DiegoCAUSA
| | - Wesley L. McKeithan
- Department of MedicineCardiovascular InstituteStanford UniversityStanfordCAUSA
| | - Mark Mercola
- Department of MedicineCardiovascular InstituteStanford UniversityStanfordCAUSA
| | | |
Collapse
|
38
|
Eugenol interacts with cardiac sodium channel and reduces heart excitability and arrhythmias. Life Sci 2021; 282:119761. [PMID: 34217764 DOI: 10.1016/j.lfs.2021.119761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/14/2021] [Accepted: 06/24/2021] [Indexed: 11/22/2022]
Abstract
AIMS Eugenol is a natural compound found in the essential oils of many aromatic plants. The compound is used as a local anesthetic because of its inhibitory effect on the voltage-gated Na+ channels (Nav), which are expressed in the nociceptive neurons. Eugenol has shown wide range of activities in the cardiovascular system; most of these activities are attributed to the modulation of voltage-sensitive Ca2+ channels. However, its action on Nav1.5, the main subtype of Nav expressed in the mammalian myocardium, is unknown. The interaction of eugenol with Nav1.5 could also contribute to its antiarrhythmic properties in vitro and ex vivo. We investigated the compound's effect on sodium current (INa) and its possible cardiac antiarrhythmic activity. METHODS The effect of eugenol on cardiac contractility was investigated using isolated atrium from guinea pig (for isometric force measurements). The compound's effect on INa was evaluated using human embryonic cell transiently expressing human Nav1.5 and patch-clamp technique. KEY FINDINGS Eugenol caused negative inotropic and chronotropic effects in the atria. In the ex vivo arrhythmia model, eugenol decreased atrial pacing disturbance induced by ouabain. Eugenol reduced the INa in a concentration-dependent manner. Furthermore, the compound left-shifted the stationary inactivation curve, delayed recovery from inactivation of the INa, and preferentially blocked the channel in the inactivated state. Importantly, eugenol was able to attenuate the late sodium current. All these aspects are considered to be antiarrhythmic. SIGNIFICANCE Overall, our findings demonstrate that eugenol has antiarrhythmic activity due, at least in part, to its interaction with Nav1.5.
Collapse
|
39
|
Farag NE, El-Kherbetawy MK, Ismail HM, Abdelrady AM, Toraih EA, Abdelbasset WK, Lashine RM, EL-dosoky M, Abed SY, Ibraheem KM, Fawzy MS, Zaitone SA. Differential Effect of Three Macrolide Antibiotics on Cardiac Pathology and Electrophysiology in a Myocardial Infarction Rat Model: Influence on Sodium Nav1.5 Channel Expression. Pharmaceuticals (Basel) 2021; 14:597. [PMID: 34206182 PMCID: PMC8308720 DOI: 10.3390/ph14070597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022] Open
Abstract
Macrolides were reported to have cardiotoxic effects presented mainly by electrocardiogram (ECG) changes with increased risk in cardiac patients. We aimed to determine the impact of three macrolides, azithromycin, clarithromycin and erythromycin, on cardiac electrophysiology, cardiac enzyme activities, histopathological changes, and sodium voltage-gated alpha subunit 5 (Nav1.5) channel expression. We used eight experimental groups of male albino rats: vehicle, azithromycin (100 mg/kg), clarithromycin (100 mg/kg), erythromycin (100 mg/kg), MI + vehicle, MI + azithromycin (100 mg/kg), MI + clarithromycin (100 mg/kg) and MI + erythromycin (100 mg/kg); each group received chronic oral doses of the vehicle/drugs for seven weeks. ECG abnormalities and elevated serum cardiac enzymes were observed particularly in rats with AMI compared to healthy rats. Microscopic examination revealed elevated pathology scores for rats treated with clarithromycin in both experiments following treatment with erythromycin in healthy rats. Although rats with MI did not show further elevations in fibrosis score on treatment with macrolides, they produced significant fibrosis in healthy rats. Downregulation of cardiac Nav1.5 transcript was observed following macrolides treatment in both groups (healthy rats and rats with MI). In conclusion, the current findings suggested the potential cardiotoxic effects of chronic doses of macrolide antibiotics in rats with MI as manifested by abnormal ECG changes and pathological findings in addition to downregulation of Nav1.5 channels. Furthermore, in the current dose ranges, azithromycin produced the least toxicity compared to clarithromycin and erythromycin.
Collapse
Affiliation(s)
- Noha E. Farag
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Department of Physiology, College of Medicine, Taif University, Taif 21974, Saudi Arabia
| | | | - Hussein M. Ismail
- Department of Cardiology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | | | - Eman A. Toraih
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA;
- Genetics Unit, Histology and Cell Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 16278, Saudi Arabia;
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza 12613, Egypt
| | - Rehab M. Lashine
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Mohammed EL-dosoky
- Department of Neuroscience Technology, College of Applied Medical Science in Jubail, Imam Abdulrahman Bin Faisal University, Jubail 35816, Saudi Arabia;
| | - Sally Yussef Abed
- Department of Respiratory Care, College of Applied Medical Science in Jubail, Imam Abdulrahman Bin Faisal University, Jubail 35816, Saudi Arabia;
| | - Khalid M. Ibraheem
- Department of Anesthesia Technology, College of Applied Medical Sciences in Jubail, Imam Abdulrahman Bin Faisal University, Jubail 35816, Saudi Arabia;
| | - Manal S. Fawzy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 1321, Saudi Arabia
| | - Sawsan A. Zaitone
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 714, Saudi Arabia
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
40
|
Joviano-Santos JV, Santos-Miranda A, Neri EA, Fonseca-Alaniz MH, Krieger JE, Pereira AC, Roman-Campos D. SCN5A compound heterozygosity mutation in Brugada syndrome: Functional consequences and the implication for pharmacological treatment. Life Sci 2021; 278:119646. [PMID: 34048814 DOI: 10.1016/j.lfs.2021.119646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 11/28/2022]
Abstract
AIMS SCN5A gene encodes the α-subunit of Nav1.5, mainly found in the human heart. SCN5A variants are the most common genetic alterations associated with Brugada syndrome (BrS). In rare cases, compound heterozygosity is observed; however, its functional consequences are poorly understood. We aimed to analyze the functional impact of de novo Nav1.5 mutations in compound heterozygosity in distinct alleles (G400R and T1461S positions) previously found in a patient with BrS. Moreover, we evaluated the potential benefits of quinidine to improve the phenotype of mutant Na+ channels in vitro. MATERIALS AND METHODS The functional properties of human wild-type and Nav1.5 variants were evaluated using whole-cell patch-clamp and immunofluorescence techniques in transiently expressed human embryonic kidney (HEK293) cells. KEY FINDINGS Both variants occur in the highly conservative positions of SCN5A. Although all variants were expressed in the cell membrane, a significant reduction in the Na+ current density (except for G400R alone, which was undetected) was observed along with abnormal biophysical properties, once the variants were expressed in homozygosis and heterozygosis. Interestingly, the incubation of transfected cells with quinidine partially rescued the biophysical properties of the mutant Na+ channel. SIGNIFICANCE De novo compound heterozygosis mutations in SNC5A disrupt the Na+ macroscopic current. Quinidine could partially reverse the in vitro loss-of-function phenotype of Na+ current. Thus, our data provide, for the first time, a detailed biophysical characterization of dysfunctional Na+ channels linked to compound heterozygosity in BrS as well as the benefits of the pharmacological treatment using quinidine on the biophysical properties of Nav1.5.
Collapse
Affiliation(s)
- J V Joviano-Santos
- Laboratory of CardioBiology, Department of Biophysics, Federal University of São Paulo, Brazil
| | - A Santos-Miranda
- Laboratory of CardioBiology, Department of Biophysics, Federal University of São Paulo, Brazil
| | - E A Neri
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, Brazil
| | - M H Fonseca-Alaniz
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, Brazil
| | - J E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, Brazil
| | - A C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, Brazil
| | - D Roman-Campos
- Laboratory of CardioBiology, Department of Biophysics, Federal University of São Paulo, Brazil.
| |
Collapse
|
41
|
Kang PW, Chakouri N, Diaz J, Tomaselli GF, Yue DT, Ben-Johny M. Elementary mechanisms of calmodulin regulation of Na V1.5 producing divergent arrhythmogenic phenotypes. Proc Natl Acad Sci U S A 2021; 118:e2025085118. [PMID: 34021086 PMCID: PMC8166197 DOI: 10.1073/pnas.2025085118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In cardiomyocytes, NaV1.5 channels mediate initiation and fast propagation of action potentials. The Ca2+-binding protein calmodulin (CaM) serves as a de facto subunit of NaV1.5. Genetic studies and atomic structures suggest that this interaction is pathophysiologically critical, as human mutations within the NaV1.5 carboxy-terminus that disrupt CaM binding are linked to distinct forms of life-threatening arrhythmias, including long QT syndrome 3, a "gain-of-function" defect, and Brugada syndrome, a "loss-of-function" phenotype. Yet, how a common disruption in CaM binding engenders divergent effects on NaV1.5 gating is not fully understood, though vital for elucidating arrhythmogenic mechanisms and for developing new therapies. Here, using extensive single-channel analysis, we find that the disruption of Ca2+-free CaM preassociation with NaV1.5 exerts two disparate effects: 1) a decrease in the peak open probability and 2) an increase in persistent NaV openings. Mechanistically, these effects arise from a CaM-dependent switch in the NaV inactivation mechanism. Specifically, CaM-bound channels preferentially inactivate from the open state, while those devoid of CaM exhibit enhanced closed-state inactivation. Further enriching this scheme, for certain mutant NaV1.5, local Ca2+ fluctuations elicit a rapid recruitment of CaM that reverses the increase in persistent Na current, a factor that may promote beat-to-beat variability in late Na current. In all, these findings identify the elementary mechanism of CaM regulation of NaV1.5 and, in so doing, unravel a noncanonical role for CaM in tuning ion channel gating. Furthermore, our results furnish an in-depth molecular framework for understanding complex arrhythmogenic phenotypes of NaV1.5 channelopathies.
Collapse
Affiliation(s)
- Po Wei Kang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218;
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Nourdine Chakouri
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032
| | - Johanna Diaz
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032
| | - Gordon F Tomaselli
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
| | - David T Yue
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Manu Ben-Johny
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218;
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032
| |
Collapse
|
42
|
Sun S, Jia Q, Zenova AY, Lin S, Hussainkhel A, Mezeyova J, Chang E, Goodchild SJ, Xie Z, Lindgren A, de Boer G, Kwan R, Khakh K, Sojo L, Bichler P, Johnson JP, Empfield JR, Cohen CJ, Dehnhardt CM, Dean R. Identification of aryl sulfonamides as novel and potent inhibitors of Na V1.5. Bioorg Med Chem Lett 2021; 45:128133. [PMID: 34044121 DOI: 10.1016/j.bmcl.2021.128133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/11/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022]
Abstract
We describe the synthesis and biological evaluation of a series of novel aryl sulfonamides that exhibit potent inhibition of NaV1.5. Unlike local anesthetics that are currently used for treatment of Long QT Syndrome 3 (LQT-3), the most potent compound (-)-6 in this series shows high selectivity over hERG and other cardiac ion channels and has a low brain to plasma ratio to minimize CNS side effects. Compound (-)-6 is also effective inshortening prolonged action potential durations (APDs) in a pharmacological model of LQT-3 syndrome in pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Unlike most aryl sulfonamide NaV inhibitors that bind to the channel voltage sensors, these NaV1.5 inhibitors bind to the local anesthetic binding site in the central pore of the channel.
Collapse
Affiliation(s)
- Shaoyi Sun
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada.
| | - Qi Jia
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Alla Y Zenova
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Sophia Lin
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Angela Hussainkhel
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Janette Mezeyova
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Elaine Chang
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Samuel J Goodchild
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Zhiwei Xie
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Andrea Lindgren
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Gina de Boer
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Rainbow Kwan
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Kuldip Khakh
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Luis Sojo
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Paul Bichler
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - J P Johnson
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - James R Empfield
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Charles J Cohen
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | | | - Richard Dean
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| |
Collapse
|
43
|
Cashman JR, Ryan D, McKeithan WL, Okolotowicz K, Gomez-Galeno J, Johnson M, Sampson KJ, Kass RS, Pezhouman A, Karagueuzian HS, Mercola M. Antiarrhythmic Hit to Lead Refinement in a Dish Using Patient-Derived iPSC Cardiomyocytes. J Med Chem 2021; 64:5384-5403. [PMID: 33942619 DOI: 10.1021/acs.jmedchem.0c01545] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Ventricular cardiac arrhythmia (VA) arises in acquired or congenital heart disease. Long QT syndrome type-3 (LQT3) is a congenital form of VA caused by cardiac sodium channel (INaL) SCN5A mutations that prolongs cardiac action potential (AP) and enhances INaL current. Mexiletine inhibits INaL and shortens the QT interval in LQT3 patients. Above therapeutic doses, mexiletine prolongs the cardiac AP. We explored structure-activity relationships (SAR) for AP shortening and prolongation using dynamic medicinal chemistry and AP kinetics in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Using patient-derived LQT3 and healthy hiPSC-CMs, we resolved distinct SAR for AP shortening and prolongation effects in mexiletine analogues and synthesized new analogues with enhanced potency and selectivity for INaL. This resulted in compounds with decreased AP prolongation effects, increased metabolic stability, increased INaL selectivity, and decreased avidity for the potassium channel. This study highlights using hiPSC-CMs to guide medicinal chemistry and "drug development in a dish".
Collapse
Affiliation(s)
- John R Cashman
- Human BioMolecular Research Institute, San Diego, California 92121, United States
| | - Daniel Ryan
- Human BioMolecular Research Institute, San Diego, California 92121, United States
| | - Wesley L McKeithan
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, California 94305, United States.,Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, San Diego, California 92037, United States
| | - Karl Okolotowicz
- Human BioMolecular Research Institute, San Diego, California 92121, United States
| | - Jorge Gomez-Galeno
- Human BioMolecular Research Institute, San Diego, California 92121, United States
| | - Mark Johnson
- Human BioMolecular Research Institute, San Diego, California 92121, United States
| | - Kevin J Sampson
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, United States
| | - Robert S Kass
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, United States
| | - Arash Pezhouman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Hrayr S Karagueuzian
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Mark Mercola
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, California 94305, United States.,Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, San Diego, California 92037, United States
| |
Collapse
|
44
|
Nakajima T, Tamura S, Kurabayashi M, Kaneko Y. Towards Mutation-Specific Precision Medicine in Atypical Clinical Phenotypes of Inherited Arrhythmia Syndromes. Int J Mol Sci 2021; 22:ijms22083930. [PMID: 33920294 PMCID: PMC8069124 DOI: 10.3390/ijms22083930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/08/2021] [Indexed: 12/19/2022] Open
Abstract
Most causal genes for inherited arrhythmia syndromes (IASs) encode cardiac ion channel-related proteins. Genotype-phenotype studies and functional analyses of mutant genes, using heterologous expression systems and animal models, have revealed the pathophysiology of IASs and enabled, in part, the establishment of causal gene-specific precision medicine. Additionally, the utilization of induced pluripotent stem cell (iPSC) technology have provided further insights into the pathophysiology of IASs and novel promising therapeutic strategies, especially in long QT syndrome. It is now known that there are atypical clinical phenotypes of IASs associated with specific mutations that have unique electrophysiological properties, which raises a possibility of mutation-specific precision medicine. In particular, patients with Brugada syndrome harboring an SCN5A R1632C mutation exhibit exercise-induced cardiac events, which may be caused by a marked activity-dependent loss of R1632C-Nav1.5 availability due to a marked delay of recovery from inactivation. This suggests that the use of isoproterenol should be avoided. Conversely, the efficacy of β-blocker needs to be examined. Patients harboring a KCND3 V392I mutation exhibit both cardiac (early repolarization syndrome and paroxysmal atrial fibrillation) and cerebral (epilepsy) phenotypes, which may be associated with a unique mixed electrophysiological property of V392I-Kv4.3. Since the epileptic phenotype appears to manifest prior to cardiac events in this mutation carrier, identifying KCND3 mutations in patients with epilepsy and providing optimal therapy will help prevent sudden unexpected death in epilepsy. Further studies using the iPSC technology may provide novel insights into the pathophysiology of atypical clinical phenotypes of IASs and the development of mutation-specific precision medicine.
Collapse
|
45
|
Li Z, Jin X, Wu T, Huang G, Wu K, Lei J, Pan X, Yan N. Structural Basis for Pore Blockade of the Human Cardiac Sodium Channel Na v 1.5 by the Antiarrhythmic Drug Quinidine*. Angew Chem Int Ed Engl 2021; 60:11474-11480. [PMID: 33684260 DOI: 10.1002/anie.202102196] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Indexed: 12/19/2022]
Abstract
Nav 1.5, the primary voltage-gated Na+ (Nav ) channel in heart, is a major target for class I antiarrhythmic agents. Here we present the cryo-EM structure of full-length human Nav 1.5 bound to quinidine, a class Ia antiarrhythmic drug, at 3.3 Å resolution. Quinidine is positioned right beneath the selectivity filter in the pore domain and coordinated by residues from repeats I, III, and IV. Pore blockade by quinidine is achieved through both direct obstruction of the ion permeation path and induced rotation of an invariant Tyr residue that tightens the intracellular gate. Structural comparison with a truncated rat Nav 1.5 in the presence of flecainide, a class Ic agent, reveals distinct binding poses for the two antiarrhythmics within the pore domain. Our work reported here, along with previous studies, reveals the molecular basis for the mechanism of action of class I antiarrhythmic drugs.
Collapse
Affiliation(s)
- Zhangqiang Li
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xueqin Jin
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Tong Wu
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Gaoxingyu Huang
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang Province, China
| | - Kun Wu
- Medical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jianlin Lei
- Technology Center for Protein Sciences, Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaojing Pan
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Nieng Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| |
Collapse
|
46
|
Li Z, Jin X, Wu T, Huang G, Wu K, Lei J, Pan X, Yan N. Structural Basis for Pore Blockade of the Human Cardiac Sodium Channel Na
v
1.5 by the Antiarrhythmic Drug Quinidine**. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Zhangqiang Li
- State Key Laboratory of Membrane Biology Beijing Advanced Innovation Center for Structural Biology Tsinghua-Peking Joint Center for Life Science School of Life Sciences Tsinghua University Beijing 100084 China
| | - Xueqin Jin
- State Key Laboratory of Membrane Biology Beijing Advanced Innovation Center for Structural Biology Tsinghua-Peking Joint Center for Life Science School of Life Sciences Tsinghua University Beijing 100084 China
| | - Tong Wu
- State Key Laboratory of Membrane Biology Beijing Advanced Innovation Center for Structural Biology Tsinghua-Peking Joint Center for Life Science School of Life Sciences Tsinghua University Beijing 100084 China
| | - Gaoxingyu Huang
- Key Laboratory of Structural Biology of Zhejiang Province Institute of Biology, Westlake Institute for Advanced Study School of Life Sciences Westlake University Hangzhou 310024 Zhejiang Province China
| | - Kun Wu
- Medical Research Center Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation Beijing Chao-Yang Hospital Capital Medical University Beijing 100020 China
| | - Jianlin Lei
- Technology Center for Protein Sciences Ministry of Education Key Laboratory of Protein Sciences School of Life Sciences Tsinghua University Beijing 100084 China
| | - Xiaojing Pan
- State Key Laboratory of Membrane Biology Beijing Advanced Innovation Center for Structural Biology Tsinghua-Peking Joint Center for Life Science School of Life Sciences Tsinghua University Beijing 100084 China
| | - Nieng Yan
- Department of Molecular Biology Princeton University Princeton NJ 08544 USA
| |
Collapse
|
47
|
Plant LD, Xiong D, Romero J, Dai H, Goldstein SAN. Hypoxia Produces Pro-arrhythmic Late Sodium Current in Cardiac Myocytes by SUMOylation of Na V1.5 Channels. Cell Rep 2021; 30:2225-2236.e4. [PMID: 32075761 PMCID: PMC7054841 DOI: 10.1016/j.celrep.2020.01.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 12/03/2019] [Accepted: 01/07/2020] [Indexed: 01/08/2023] Open
Abstract
Acute cardiac hypoxia produces life-threatening elevations in late sodium current (ILATE) in the human heart. Here, we show the underlying mechanism: hypoxia induces rapid SUMOylation of NaV1.5 channels so they reopen when normally inactive, late in the action potential. NaV1.5 is SUMOylated only on lysine 442, and the mutation of that residue, or application of a deSUMOylating enzyme, prevents hypoxic reopenings. The time course of SUMOylation of single channels in response to hypoxia coincides with the increase in ILATE, a reaction that is complete in under 100 s. In human cardiac myocytes derived from pluripotent stem cells, hypoxia-induced ILATE is confirmed to be SUMO-dependent and to produce action potential prolongation, the pro-arrhythmic change observed in patients. The cardiac channel NaV1.5 passes pro-arrhythmic late sodium currents in response to hypoxia. Plant et al. demonstrate the pathophysiological mechanism to be rapid, hypoxia-induced monoSUMOylation of NaV1.5 channels. Blocking SUMOylation of lysine442 prevents hypoxia-induced late currents and attendant prolongation of the action potential in human cardiomyocytes derived from pluripotent stem cells.
Collapse
Affiliation(s)
- Leigh D Plant
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Dazhi Xiong
- Departments of Pediatrics and Physiology & Biophysics, University of California, Irvine, 1001 Health Sciences Road, Irvine Hall, Irvine, CA 92697, USA
| | - Jesus Romero
- Departments of Pediatrics and Physiology & Biophysics, University of California, Irvine, 1001 Health Sciences Road, Irvine Hall, Irvine, CA 92697, USA
| | - Hui Dai
- Departments of Pediatrics and Physiology & Biophysics, University of California, Irvine, 1001 Health Sciences Road, Irvine Hall, Irvine, CA 92697, USA
| | - Steve A N Goldstein
- Departments of Pediatrics and Physiology & Biophysics, University of California, Irvine, 1001 Health Sciences Road, Irvine Hall, Irvine, CA 92697, USA.
| |
Collapse
|
48
|
Abe I, Wang P, Takahashi M, Ohno S, Ono K, Takahashi N. Familial sick sinus syndrome possibly associated with novel SCN5A mutation diagnosed in pregnancy. HeartRhythm Case Rep 2021; 7:117-122. [PMID: 33665115 PMCID: PMC7897738 DOI: 10.1016/j.hrcr.2020.11.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Ichitaro Abe
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University, Oita, Japan
| | - Pu Wang
- Department of Pathophysiology, Faculty of Medicine, Oita University, Oita, Japan.,Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Masaki Takahashi
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University, Oita, Japan
| | - Seiko Ohno
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Katsushige Ono
- Department of Pathophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Naohiko Takahashi
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University, Oita, Japan
| |
Collapse
|
49
|
Plumereau Q, Theriault O, Pouliot V, Moreau A, Morel E, Fressart V, Denjoy I, Delinière A, Bessière F, Chevalier P, Gamal El-Din TM, Chahine M. Novel G1481V and Q1491H SCN5A Mutations Linked to Long QT Syndrome Destabilize the Nav1.5 Inactivation State. CJC Open 2021; 3:256-266. [PMID: 33778442 PMCID: PMC7984979 DOI: 10.1016/j.cjco.2020.09.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 09/27/2020] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Nav1.5, which is encoded by the SCN5A gene, is the predominant voltage-gated Na+ channel in the heart. Several mutations of this gene have been identified and reported to be involved in several cardiac rhythm disorders, including type 3 long QT interval syndrome, that can cause sudden cardiac death. We analyzed the biophysical properties of 2 novel variants of the Nav1.5 channel (Q1491H and G1481V) detected in 5- and 12-week-old infants diagnosed with a prolonged QT interval. METHODS The Nav1.5 wild-type and the Q1491H and G1481V mutant channels were reproduced in vi tr o. Wild-type or mutant channels were cotransfected in human embryonic kidney (HEK) 293 cells with the beta 1 regulatory subunit. Na+ currents were recorded using the whole-cell configuration of the patch-clamp technique. RESULTS The Q1491H mutant channel exhibited a lower current density, a persistent Na+ current, an enhanced window current due to a +20-mV shift of steady-state inactivation, a +10-mV shift of steady-state activation, a faster onset of slow inactivation, and a recovery from fast inactivation with fast and slow time constants of recovery. The G1481V mutant channel exhibited an increase in current density and a +7-mV shift of steady-state inactivation. The observed defects are characteristic of gain-of-function mutations typical of type 3 long QT interval syndrome. CONCLUSIONS The 5- and 12-week-old infants displayed prolonged QT intervals. Our analyses of the Q1491H and G1481V mutations correlated with the clinical diagnosis. The observed biophysical dysfunctions associated with both mutations were most likely responsible for the sudden deaths of the 2 infants.
Collapse
Affiliation(s)
| | | | | | - Adrien Moreau
- Inserm U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France
| | - Elodie Morel
- Lyon Reference Center for Inherited Arrhythmias, Louis Pradel Cardiovascular Hospital, Bron, France
| | - Véronique Fressart
- Centre de Génétique Moléculaire et Chromosomique, Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Antoine Delinière
- Lyon Reference Center for Inherited Arrhythmias, Louis Pradel Cardiovascular Hospital, Bron, France
| | - Francis Bessière
- Lyon Reference Center for Inherited Arrhythmias, Louis Pradel Cardiovascular Hospital, Bron, France
| | - Philippe Chevalier
- Lyon Reference Center for Inherited Arrhythmias, Louis Pradel Cardiovascular Hospital, Bron, France
- Department of Rhythmology, Louis Pradel Cardiovascular Hospital, Lyon, France
- Université de Lyon, Lyon, France
| | | | - Mohamed Chahine
- CERVO Brain Research Center, Quebec City, Quebec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
50
|
Föhr KJ, Nastos A, Fauler M, Zimmer T, Jungwirth B, Messerer DAC. Block of Voltage-Gated Sodium Channels by Atomoxetine in a State- and Use-dependent Manner. Front Pharmacol 2021; 12:622489. [PMID: 33732157 PMCID: PMC7959846 DOI: 10.3389/fphar.2021.622489] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Atomoxetine, a neuroactive drug, is approved for the treatment of attention-deficit/hyperactivity disorder (ADHD). It is primarily known as a high affinity blocker of the noradrenaline transporter, whereby its application leads to an increased level of the corresponding neurotransmitter in different brain regions. However, the concentrations used to obtain clinical effects are much higher than those which are required to block the transporter system. Thus, off-target effects are likely to occur. In this way, we previously identified atomoxetine as blocker of NMDA receptors. As many psychotropic drugs give rise to sudden death of cardiac origin, we now tested the hypothesis whether atomoxetine also interacts with voltage-gated sodium channels of heart muscle type in clinically relevant concentrations. Electrophysiological experiments were performed by means of the patch-clamp technique at human heart muscle sodium channels (hNav1.5) heterogeneously expressed in human embryonic kidney cells. Atomoxetine inhibited sodium channels in a state- and use-dependent manner. Atomoxetine had only a weak affinity for the resting state of the hNav1.5 (Kr: ∼ 120 µM). The efficacy of atomoxetine strongly increased with membrane depolarization, indicating that the inactivated state is an important target. A hallmark of this drug was its slow interaction. By use of different experimental settings, we concluded that the interaction occurs with the slow inactivated state as well as by slow kinetics with the fast-inactivated state. Half-maximal effective concentrations (2–3 µM) were well within the concentration range found in plasma of treated patients. Atomoxetine also interacted with the open channel. However, the interaction was not fast enough to accelerate the time constant of fast inactivation. Nevertheless, when using the inactivation-deficient hNav1.5_I408W_L409C_A410W mutant, we found that the persistent late current was blocked half maximal at about 3 µM atomoxetine. The interaction most probably occurred via the local anesthetic binding site. Atomoxetine inhibited sodium channels at a similar concentration as it is used for the treatment of ADHD. Due to its slow interaction and by inhibiting the late current, it potentially exerts antiarrhythmic properties.
Collapse
Affiliation(s)
- Karl Josef Föhr
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm, Germany
| | - Ariadni Nastos
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm, Germany
| | - Michael Fauler
- Department of General Physiology, Ulm University, Ulm, Germany
| | - Thomas Zimmer
- Institute of Physiology, University Hospital Jena, Jena, Germany
| | - Bettina Jungwirth
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm, Germany
| | | |
Collapse
|